Sex Differences in Cardiovascular Impact of Early Metabolic Impairment: Interplay between Dysbiosis and Adipose Inflammation ============================================================================================================================ * Haneen S. Dwaib * Ibrahim AlZaim * Ghina Ajouz * Ali H. Eid * Ahmed El-Yazbi ## Visual Overview ![Figure1](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F1.medium.gif) [Figure1](http://molpharm.aspetjournals.org/content/102/1/60/F1) ## Abstract The evolving view of gut microbiota has shifted toward describing the colonic flora as a dynamic organ in continuous interaction with systemic physiologic processes. Alterations of the normal gut bacterial profile, known as dysbiosis, has been linked to a wide array of pathologies. Of particular interest is the cardiovascular–metabolic disease continuum originating from positive energy intake and high-fat diets. Accumulating evidence suggests a role for sex hormones in modulating the gut microbiome community. Such a role provides an additional layer of modulation of the early inflammatory changes culminating in negative metabolic and cardiovascular outcomes. In this review, we will shed the light on the role of sex hormones in cardiovascular dysfunction mediated by high-fat diet-induced dysbiosis, together with the possible involvement of insulin resistance and adipose tissue inflammation. Insights into novel therapeutic interventions will be discussed as well. **SIGNIFICANCE STATEMENT** Increasing evidence implicates a role for dysbiosis in the cardiovascular complications of metabolic dysfunction. This minireview summarizes the available data on the sex-based differences in gut microbiota alterations associated with dietary patterns leading to metabolic impairment. A role for a differential impact of adipose tissue inflammation across sexes in mediating the cardiovascular detrimental phenotype following diet-induced dysbiosis is proposed. Better understanding of this pathway will help introduce early approaches to mitigate cardiovascular deterioration in metabolic disease. ## Introduction The gut microbiota (GM) is a complex ecosystem that can be described as a dynamic organ with an active role in human health and disease (Putignani et al., 2014). The microbial community has high plasticity and is sensitive to several stimuli including environmental, hormonal, dietary, and stress-related factors (Putignani et al., 2014). Nevertheless, diet remains one of the most vigorous modulators of GM (David et al., 2014) with Western-type calorie-dense diets driving an imbalance of microorganisms in the gut or dysbiosis (Sen et al., 2017). This is particularly relevant to the steady rise in the prevalence of metabolic disease like diabetes, obesity, and their complications, driven by increased caloric intake after the global shift to the Western diets, rich in saturated fat and refined sugars (Lutsey et al., 2008; Misra et al., 2010). As such, there has been an increasing interest in studying dysbiosis in these maladies together with the impact of its modification as a therapeutic option. Significantly, most of the health burden associated with metabolic dysfunction is due to the high risk of cardiovascular mortality and morbidity due to ischemic heart disease, ischemic stroke, cardiac metabolic dysfunction, and heart failure (Ash-Bernal and Peterson, 2006; von Bibra et al., 2016). Of note, cardiovascular risk evoked by metabolic impairment has long been associated with a state of chronic low-grade inflammation (de Rooij et al., 2009). Indeed, under circumstances leading to dysbiosis, GM can contribute to this inflammatory state. Normally, the host health/gut bacteria interaction occurs through exposure to either bacterial components known as pathogen-associated molecular patterns, like flagella and cell wall constituents like lipopolysaccharide (LPS) (Tilg et al., 2019) or to the metabolites produced by bacterial digestion and processing of ingested food, which were shown to have several effects including modulation of the function of immune and autonomic nervous system as will be discussed below. Therefore, dysbiosis outcomes depend on the changes in bacterial phyla residing in the gut. Interestingly, considerable sex-dependent differences were reported in inflammatory changes and cardiovascular risk associated with metabolic dysfunction. Recent literature shows that metabolic impairment in humans leads to different inflammatory profiles across sexes with increased production of proinflammatory cytokines in males (ter Horst et al., 2020). Indeed, premenopausal females are less prone to adverse cardiovascular events (Mosca et al., 2011), and varying cardiovascular profiles secondary to metabolic deterioration are observed in either sex (Gerdts and Regitz-Zagrosek, 2019). Although sex-dependent differences in metabolic-derived cardiovascular diseases (CVDs) are typically attributed to estrogen-driven alteration in insulin sensitivity, adiposity, adipocyte size and function, as well as adipose tissue (AT) susceptibility to inflammation (Ribas et al., 2010; Pradhan, 2014; Zore et al., 2018), sex-dependent differences in GM together with its vulnerability to dysbiosis add a new layer of complexity to the paradigm. GM appears to play an important role in mediating the differential patterns observed in diet-induced metabolic and cardiovascular dysfunction across sexes. Although the exact mechanism has yet to be comprehensively and systematically investigated, we attempt here to shed the light on the potential mechanisms through which dysbiosis mediates cardiovascular dysfunction in early metabolic impairment in a sex-dependent manner. We explore the sex differences in high-fat diet- (HFD) induced dysbiosis and the consequent AT inflammatory changes and cardiovascular dysfunction in the context of early metabolic deterioration. As well, we summarize some of the available evidence regarding possible therapeutic interventions to address these disorders via targeting the gut microbiome homeostasis. ## Sex-Dependent Differences in Gut Microbiota in the Healthy State The assumption of dysbiosis in disease states necessitates a fundamental knowledge of the composition and function of GM in healthy individuals. Nevertheless, a unified healthy GM profile has not been defined at any profound taxonomic resolution owing to several endogenous and exogenous factors. These include interindividual host genetic and environmental differences (Hooper et al., 2001; Abdul-Aziz et al., 2016; Rothschild et al., 2018), disparate GM growth rates, strain-level diversities, and variants within microbial genes (Huttenhower et al., 2012; Korem et al., 2015; Zeevi et al., 2019). However, high taxonomic diversity, along with high microbial gene richness and stable microbiome functions represent characteristics of a healthy GM (Huttenhower et al., 2012). The major bacterial phyla inhabiting the human gut are *Fermicutes*, *Bacteroidetes*, *Proteobacteria*, *Verrucomicrobia*, *Actinobacteria*, and *Fusobacteria*, with *Firmicutes* and *Bacteroidetes* accounting for almost 70% of the total microbiota (Zoetendal et al., 2008; Mariat et al., 2009) and their *Firmicutes*/*Bacteroidetes* (*F*/B) ratio changing under situations of metabolic impairment (Turnbaugh et al., 2006). The homeostatic state in which the GM is healthy and balanced is referred to as a state of eubiosis (Iebba et al., 2016). Significantly, several studies highlighted sex-dependent variations of the GM in health and disease (Org et al., 2016; Han et al., 2017; Razavi et al., 2019). It was shown that *Drosophila* melanogaster strains exhibit a differential abundance of microbes across sexes irrespective of nutritional conditions (Han et al., 2017). Interestingly, similar results were also obtained in different strains of mice where the abundance of several taxa exhibited significant sex-dependent differences (Org et al., 2016). Furthermore, such differences were also evident in mice fed either normal chow or HFD, suggesting sex-by-diet interactions (Org et al., 2016). Indeed, the impact of gonadectomy and sex hormone replacement on GM is far from settled. This is of particular interest in light of the reduced susceptibility of premenopausal women to metabolic and cardiometabolic diseases (Santos-Marcos et al., 2019) where numerous lines of evidence suggested a role for female sex hormones in enhancing the diversity of the GM (Song et al., 2020). A cross-sectional study revealed that although sex-dependent differences in GM were observed before puberty, they tended to increase after puberty with significant differences in *β*-diversity (interindividual dissimilarity) but not in *α*-diversity (intraindividual bacterial diversity) (Yuan et al., 2020). Furthermore, studies have shown a stronger impact of female gonadal hormones on GM. A study in rats showed that although the sex-dependent differences in GM community persisted after gonadectomy, the detrimental impact was more pronounced in female rats, especially when animals were overfed (Santos-Marcos et al., 2020). This is indeed in line with observations in humans demonstrating a shift in the GM profile in postmenopausal women compared with age matched men (Santos-Marcos et al., 2018). Along the same lines, the strong associations observed between sex and *α*-diversity in young adults, although persisted after adjusting for cardiometabolic parameters, tended to diminish after 40 years of age (de la Cuesta-Zuluaga et al., 2019), consistent with an age-related decline in the level of sex hormones. Importantly, the interaction between GM and female sex hormones appears to be bidirectional, whereby a study on rats showed that several GM-derived microRNAs were reported to modulate steroid biosynthesis and estrogen signaling (Santos-Marcos et al., 2020). Although less clear in human studies, accumulating evidence from experiments on mice suggests that the differential diversity in GM can drive sexually dimorphic immune responses (Elderman et al., 2018; Felix et al., 2018). It was even suggested that sexual dimorphism in susceptibility to certain autoimmune disorders, like Type 1 diabetes mellitus, was mediated by GM in rodent models, and the alteration of GM at an early age may protect against genetic predisposition to autoimmune diseases (Markle et al., 2013; Yurkovetskiy et al., 2013; Candon et al., 2015). This sex-dependent dysbiosis in disease prognosis was suggested to mediate manganese-induced neurotoxicity (Chi et al., 2017). As these factors augment the complexity of the host environment-microbiota interactions, it becomes plausible that diet-induced GM alteration leading to metabolic impairment will trigger distinct inflammatory responses in either sex, culminating in disparate cardiovascular consequences. ## Sex-Dependent Gut Microbiome Alterations in Early Metabolic Impairment Early metabolic impairment has long been discussed in the literature, yet there has been no consensus on the exact definition and the diagnostic criteria. This is despite the fact that a significant proportion of the global population exhibits suboptimal metabolic health, primarily due to excessive caloric consumption and sedentary lifestyles resulting in the increased prevalence of metabolic diseases such as insulin resistance, obesity, and diabetes (Chatelier et al., 2013; Zheng et al., 2018; Jaacks et al., 2019; Frost et al., 2021). This is mirrored by an increased prevalence of metabolic dysfunction-associated cardiovascular diseases (Lakka et al., 2002). Indeed, recent research identified early stages of metabolic deterioration such as prediabetes or metabolically unhealthy normal weight as risk factors of cardiovascular disease (Stefan, 2020; Alderman, 2021). Despite the various pathologic mechanisms culminating in the emergence of these disorders, it seems that they are correlated with GM alterations referred to as dysbiosis (Qin et al., 2012; Qin et al., 2014; Allin et al., 2018). Next-generation sequencing of the gut microbiome had a major role in unfolding the involvement of GM in regulating the host metabolism. Metabolic-related dysbiosis is usually exemplified by altering the abundance of *Bacteroides*, *Prevotella*, *Desulfovibrio*, *Lactobacillus*, and *Oxalobacter* genera in the gut (Tyakht et al., 2013; Clemente et al., 2015; Smits et al., 2017). Nevertheless, the precise dynamics of GM involvement in metabolic diseases is not fully explored yet. Indeed, whether the disease-associated aberrant microbiota underpins disease causation or represents a secondary phenomenon after disease onset and progression has been widely debated (Bäckhed et al., 2004; Pedersen et al., 2016; Qin et al., 2010). Fig. 1 outlines the proposed framework linking dysbiosis in metabolic dysfunction with the pathogenesis of cardiovascular complications together with the interplay with sex-dependent factors as described below. ![Fig. 1.](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F2.medium.gif) [Fig. 1.](http://molpharm.aspetjournals.org/content/102/1/60/F2) Fig. 1. High fat diet-mediated gut dysbiosis and links to cardiovascular disease and sex-dependent factors. Significant increase in the *F*/*B* ratio occurs after the consumption of a high-fat diet. The ensuing gut microbiota dysbiosis detrimentally affects the adipose tissue and the cardiovascular system through intricate pathways. Mechanistically, gut microbiota dysbiosis enhances lipopolysaccharide production, as well as its chylomicron-mediated transport and paracellular diffusion. The latter is possible due to gut microbiota dysbiosis-mediated dysfunction of tight junctions leading to a compromised gut integrity. Locally, LPS activates, through TLR4, the proinflammatory NF-κB pathway. Systemically, the increased levels of serum LPS results in endotoxemia. Importantly, sex hormones are partly responsible for the differential modulation of these pathways in either sex as indicated. Pathways involved in gut microbiota dysbiosis are presented in red, whereas those counteracting them in black. AT; COX2, cyclooxygenase 2; ER-*β*; *F*/*B*; GM; HFD; IAP, intestinal alkaline phosphatase; IL; iNOS, inducible nitric oxide synthase; IR, insulin resistance; LPS; MetS, metabolic syndrome; MUC2; MyD88; NF-κB; PUFA, polyunsaturated fatty acid; TLR4; TNF-*α*. #### Sex-Dependent Differences in Diet-Induced Dysbiosis As diet remains the cornerstone for GM modulation in humans and animal models (David et al., 2014; Carmody et al., 2015), dysbiosis has been the focus of research on diet induced pathologies. HFD was consistently reported to provoke dysbiosis by altering the scale of the major gut phyla, increasing *F*/*B* ratio as well as an increase in *Proteobacteria,* which were ushered with impaired metabolic and cardiovascular function (Moreira et al., 2012; Murphy et al., 2015). *Bacteroidetes* is considered the most prevalent gram-negative bacteria in the gut and is essentially considered beneficial due to their capacity to modulate caloric absorption through polysaccharide metabolism (Wexler, 2007). On the other hand, *Firmicutes* are largely gram positive and are capable of producing various short-chain fatty acids (SCFAs) (Den Besten et al., 2013). It is generally accepted that a higher *F*/*B* ratio is usually observed in overweight and obese subjects (Ley et al., 2006; Million et al., 2012; Kasai et al., 2015), and a reduction in the *F*/*B* ratio has been associated with weight loss (Ley et al., 2006; Turnbaugh et al., 2006). However, the opposite was documented as well where HFD and diet-induced obesity were associated with decreased *F*/*B* ratio in human and animal models in both sexes (Collado et al., 2008; Schwiertz et al., 2010). Indeed, dysbiosis severity in different situations of metabolic impairment appeared to be sex dependent (Org et al., 2016). Premenopausal women were shown to have a higher F*/*B ratio than postmenopausal women and men (Santos-Marcos et al., 2018). Accumulating evidence suggested that women harbor a higher *F*/*B* ratio in comparison with men, even after adjusting for body mass index (BMI) (Mueller et al., 2006; Dominianni et al., 2015). In fact, *F*/*B* ratio was found to be highly influenced by BMI and has been used as an indicator of gut dysbiosis with a higher *F*/*B* ratio indicating a more pronounced dysbiotic microbiome (Kasai et al., 2015). Another human study showed that among subjects with BMI greater than 33, men exhibited a significantly lower *F*/*B* ratio in comparison with women, whereas the opposite was observed in subjects with a BMI lower than 33 as well as in postmenopausal women (Haro et al., 2016). Sex-differential dysbiosis was also observed among lean men and women, as postmenopausal women had a similar GM signature as men, whereas obesity abolished these differences. The same study emphasized the tight correlation between sex hormones and GM diversity. Premenopausal women had higher *Bifidobacterium* and lower *Bacteroides* than men and postmenopausal women, as GM community could predict testosterone level in humans and recipient mice of human fecal microbiome (Mayneris-Perxachs et al., 2020). In mechanistic terms, estrogen receptor-*β* (ER*β*) has been proposed to be a modulator of sex-dependent dysbiosis. For instance, ER*β* knockout female mice on isoflavone and fiber-rich feeding achieved a state of eubiosis with an increase in *Bacteroidetes* and a reduction in *Firmicutes* and *Proteobacteria*. However, when these mice were switched to an isocaloric low-fiber and simple-sugar rich diet, the knockout mice had more pronounced dysbiosis and reduced *Bacteroidetes* in favor of *Proteobacteria* compared with their wild-type counterparts (Menon et al., 2013), suggesting a protective effect of estrogen in diet-induced dysbiosis. Similarly, genetically obese ob/ob mice show an elevation of *F*/*B* ratio (Turnbaugh et al., 2006). Interestingly, hormonal treatment with 17*β*-estradiol (E2) in female mice was found to correct HFD-related dysbiosis in ob/ob and wild-type mice by increasing the heterogeneity of GM distribution and reducing the *F*/*B* ratio compared with the vehicle group (Acharya et al., 2019). Moreover, androgenization of young and adult ovariectomized female Wistar rats induced dysbiosis regardless of dietary intervention. It reduced GM diversity, elevated *F*/*B* ratio, and impaired overall metabolic function (Moreno-Indias et al., 2016). Although human studies link dysbiosis to obesity, results from experiments on animals implicate HFD as the culprit even in absence of obesity. This was supported with results from resistin-like molecule *β* knockout female mice, a model that lacks the specific gene for colonic goblet cells, whereas HFD-fed mice were not obese. These mice presented with the typical HFD-related dysbiosis (Hildebrandt et al., 2009). It is noteworthy that the type of fat affects the microbiome alteration as diets rich in saturated fatty acids are thought to contribute to the development of endotoxemia by enhancing the production of LPS, whereas polyunsaturated fatty acids are suggested to exert protective effects by influencing systemic endotoxin concentrations, LPS clearance, bile acid metabolism, intestinal alkaline phosphatase activity, intestinal mucosal permeability, and microbiota composition diversity (Bellenger et al., 2019; Cândido et al., 2020). Moreover, saturated fatty acids were recently demonstrated to act as a nonmicrobial toll-like receptor 4 (TLR4) agonists, triggering myeloid differentiation primary response 88 (MyD88)-dependent or independent inflammatory pathways culminating in the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-*κ*B) and the production of inflammatory cytokines, in a similar fashion to LPS (Rocha et al., 2016). Moreover, mice fed HFD rich in omega-6 fatty acids but not omega-3 fatty acids exhibited a more pronounced metabolic endotoxemia (Kaliannan et al., 2015). Transgenic mice over-converting omega-6 to omega-3 in tissues exhibited an augmented production of intestinal alkaline phosphatase, which subsequently alters GM composition, reduces LPS production, maintains gut barrier function, and reduces endotoxemia (Kaliannan et al., 2015). Importantly, linoleic acid and *α*-linolenic acid-enriched HFD-fed obese mice exhibited a sex-dependent alleviation of endotoxemia and systemic and AT inflammation, which was associated with sex-dependent alterations in GM composition (Zhuang et al., 2018). This suggests that linoleic acid may provide a protective effect against metabolic endotoxemia in female mice, whereas *α*-linolenic acid exerts a similar effect in male mice through modulating the gut-adipose tissue axis (Zhuang et al., 2018). These variabilities highlight the complex interplay of sex and diet in shaping the microbiome community and its metabolic consequences. #### Sex-Dependent Differences in Dysbiosis Consequences: Compromised Gut Integrity and Metabolic Endotoxemia As mentioned in the previous section, the detrimental effects of consumption of HFD are partly mediated by dysbiosis evidenced by an augmented and reduced relative abundance of *Proteobacteria* and *Bacteroidetes*, respectively. Disruption of gut barrier function as well as metabolic endotoxemia ensue (Satokari, 2020). Although a sizable body of evidence consistently supports an increased *F*/*B* ratio under these circumstances, an increased LPS would not be expected given the observed reduction of the gram-negative *Bacteroidetes*. Paradoxically, increased plasma LPS levels were reported after HFD in animal experiments (Cani et al., 2008) and in obese humans in comparison with their lean controls (Stoll et al., 2004; Trøseid et al., 2013). Such observations can be attributed to the reduced expression of intestinal epithelial tight junction proteins leading to a compromised gut barrier in animals and humans, allowing for increased LPS transportation (Saad et al., 2016). As well, chylomicrons, lipoprotein particles mediating intestinal fat absorption, were found to facilitate LPS transport across the intestinal lumen through enterocyte-mediated absorption (Ghoshal et al., 2009), offering an additional mechanism by which HFD can contribute to increase plasma LPS levels. Expectedly, metabolic endotoxemia will follow, whereas the transport of microbial-associated molecular patterns leads to systemic low-grade and subclinical inflammation via the activation of TLR4 (Turnbaugh et al., 2006; Cani et al., 2007; Rodriguez et al., 2020). Indeed, LPS binds to cluster of differentiation 14 and the TLR4/Myeloid differentiation factor 2 receptor complex (Kitchens and Thompson, 2005; Lu et al., 2008). TLR4 cascade was shown to mediate HFD-induced inflammation. HFD feeding led to an increased serum and fecal LPS, concurrently with increased *F*/*B* ratio in male wild-type mice. Moreover, these mice suffered from diet-induced colitis indicated by increased intestinal tumor necrosis factor *α* (TNF*α*), interleukin (IL)-1*β*, inducible nitric oxide synthase, cyclooxygenase 2, and phosphor-IK kinase *β* NF-*κ*B expression and activity together with a compromised gut integrity and reduced expression of tight junctions protein occludin and claudin-1 (Kim et al., 2012). On the other hand, HFD-fed TLR4 knockout mice were protected from metabolic endotoxemia, as they had lower serum LPS compared with their low-fat fed counterparts. At the same time, the knockout mouse intestine did not show the same HFD-induced inflammatory consequences (Kim et al., 2012). Sexual dimorphism in the response to HFD-induced gut barrier permeability and metabolic endotoxemia has been attributed to differential hormonal modulation of these pathologic processes. Estrogen has been shown to prevent metabolic endotoxemia and chronic low-grade inflammation, which was suggested to be a contributor to the cardiometabolic privilege of premenopausal women (Santos-Marcos et al., 2019). Indeed, 17*β*-estradiol-treated male, ovariectomized, and intact female mice exhibited reduced LPS production and lower susceptibility to metabolic endotoxemia and metabolic syndrome partly through upregulation of intestinal alkaline phosphatase (Kaliannan et al., 2018). On the same note, one study highlighted a protective effect of progesterone against endotoxemia, where plasma levels of LPS negatively correlated with plasma progesterone levels but positively correlated with TNF-*α* plasma levels in pregnant women (Zhou et al., 2019). This differential effect of sex hormones was shown to be due to the alteration of expression and function of tight junctions and mucin production, thus regulating HFD-induced alteration of gut permeability. For example, progesterone was found to upregulate the expression of the tight junction protein occludin and inhibit NF-*κ*B activation in LPS-stimulated Caco-2 cells (Zhou et al., 2019). Similarly, estrogens promoted gut barrier function through several mechanisms. For instance, ER*β* was proposed to play a crucial role in regulating cellular differentiation in colonic tissue as ER*β*−/− mice exhibit epithelial hyperproliferation and compromised integrity (Imamov et al., 2004; Wada-Hiraike et al., 2006). This suggests a homeostatic role of estrogen in the maintenance of colon integrity. Furthermore, estrogen-ER*β* cascade was suggested to enhance mucin 2 (MUC2) secretion by intestinal goblet cells, which offers epithelial protection. Hence, the deletion of ER*β* disrupted the colon mucin layer in female mice (Wada-Hiraike et al., 2006; Diebel et al., 2015). Likewise, female mice during proestrus stage, characterized by high estrogen levels, are protected against intestinal injury in comparison with their male counterparts and female mice in the diestrus stage that is characterized by low circulating estrogen (Homma et al., 2005; Sheth et al., 2010). This can be attributed to the lower mucus thickness observed in the colon of diestrus female mice and male mice compared with the proestrus ones (Elderman et al., 2017). Apart from the induction of MUC2 production, estrogen is also found to upregulate the expression of tight junctions in both male and ovariectomized female rodents as well as in vitro monolayer cultures (Homma et al., 2005; Braniste et al., 2009; Looijer-van Langen et al., 2011). Consistent with these observations, the differential effects of estrogen supplementation on metabolic health observed in pre- and postmenopausal women suggest that the loss of ovarian function may profoundly alter estrogen signaling (Hulley et al., 1998; Rossouw et al., 2002). Indeed, it was shown that ovariectomized mice exhibit temporal and regional changes in gastrointestinal permeability due to disruption of tight junctions (Collins et al., 2017). Intriguingly, it was shown in some studies that TLR4 level in female macrophages were higher after ovariectomy (Rettew et al., 2009). Although males tend to have higher metabolic endotoxemia, orchiectomized mice were more susceptible to endotoxemia, and isolated macrophages presented higher TLR4 level than intact males. At the same time, testosterone treatment attenuated these events, suggesting an immunosuppressive effect of testosterone (Rettew et al., 2008). These findings emphasize the complexity of the effect of sex hormones on dysbiosis outcomes. Nevertheless, female sex hormones in animal models consistently exhibited a protective effect on gut integrity and the resulting metabolic endotoxemia, mainly by upregulating intestinal tight junction proteins. #### Sex-Dependent Differences in Dysbiosis-Induced Insulin Resistance and Adipose Inflammation Diet-induced metabolic derangement and its correlated cardiovascular dysfunction have been suggested to be an outcome of early adipose inflammation and insulin resistance, even in the absence of explicit hyperglycemia (Shah et al., 2008; Nishimura et al., 2009; Gollasch, 2017; Elkhatib et al., 2019; Rafeh et al., 2020). As previously demonstrated, dysbiosis and altered gut permeability precede the emergence of metabolic syndrome (Martínez-Oca et al., 2020). Since HFD consumption increased intestinal permeability by impairing the function of tight junctions (Cani et al., 2008), intestinal hyperpermeability and GM dysbiosis were suggested to further the inflammatory phenotype in AT and to increase the risk of cardiovascular diseases (Serino et al., 2012; Kallio et al., 2015; Clemente-Postigo et al., 2019; Gasmi et al., 2021). A great body of research attempted to establish an understanding of the gut-adipose axis in metabolic dysfunction (Poggi et al., 2007; Samuel et al., 2008; Serino et al., 2012). Accordingly, white adipose tissue (WAT) was identified as a major target of GM. Not only were GM fermentation products shown to regulate WAT energy balance (Samuel et al., 2008), but also GM regulated fat deposition in AT as well as insulin resistance (Bäckhed et al., 2004; Bäckhed et al., 2007; Velagapudi et al., 2010). Such observations are of relevance given the contribution of WAT to the development of metabolic inflammation leading to insulin resistance and cardiovascular dysfunction (Bouloumié et al., 2005; Hotamisligil, 2006; Bouloumié et al., 2008). In HFD-fed diabetic mice, dysbiosis increased the stromal vascular fraction in WAT, in particular macrophages, lymphocytes, and preadipocytes (Serino et al., 2012). Additionally, LPS migration into the circulation has been suggested to be a contributing factor to the onset of AT inflammation, insulin resistance, obesity, and diabetes (Cani et al., 2007; Cani et al., 2008; Hersoug et al., 2016). Interestingly, HFD consumption had a similar effect to LPS subcutaneous infusion on elevating serum LPS and promoting AT inflammation in male mice (Cani et al., 2007). Significantly, HFD-fed TLR4 knockout mice did not have increased proinflammatory cytokines in the isolated epidydimal AT depot, whereas wild-type mice were hyperinsulinemic and exhibited a proinflammatory response in the epidydimal WAT manifested by increased TNF*α*, IL-1*β*, and IL-6, and macrophage infiltration (Kim et al., 2012). Moreover, TLR4 knockout male mice had an increased insulin sensitivity in subcutaneous and epidydimal WAT even in presence of HFD feeding (Poggi et al., 2007). Consistently, HFD-fed male mice showed an improved metabolic status after eight weeks of antibiotic treatment, as serum LPS, insulin, and fasting glucose were reduced. The epidydimal WAT in the treated group had lower TLR4 activation, Jun NH2-terminal kinase inactivation, lower nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor *α* degradation, and inhibition of insulin receptor substrate 1 Ser307 phosphorylation. Additionally, the treatment group had improved insulin signaling with increased protein kinase B (Akt) phosphorylation and reduced macrophage infiltration. It is noteworthy that HFD-fed germ-free mouse models were not vulnerable to HFD-induced insulin resistance (Fleissner et al., 2010; Rabot et al., 2010). As for sex-based differences, a recent study reported that HFD induced weight gain and insulin resistance in males but not in female mice with differences in gut microbiome recorded at baseline (Peng et al., 2020). Moreover, E2 treatment in intact female mice improved HFD-induced weight gain, glucose intolerance, and insulin resistance possibly as a result of the downregulation of lipogenic genes, such as sterol response element binding protein-1 (SREBP-1) and leptin and resistin genes expression in WAT (Bryzgalova et al., 2008). Moreover, E2 intervention in ovariectomized female mice prevented HFD-induced obesity (Bless et al., 2014). Administration of E2 in ovariectomized female rats increased leptin sensitivity and led to preferential subcutaneous adiposity, resembling the intact female littermates (Clegg et al., 2006). Notably, subcutaneous fat pads were suggested to be more sensitive to insulin than visceral fat (Chang et al., 2018). This might be one of the many protective roles of estrogens against metabolic impairment related to AT dysfunction, as premenopausal women have more subcutaneous AT than men and postmenopausal women (Lemieux et al., 1993). Furthermore, E2 was proposed to ubiquitinate and degrade HIF1-*α*, eventually reducing adipose inflammation and its subsequent metabolic derangements (Kim et al., 2014). Typically, insulin resistance is associated with hypertrophic adipose expansion leading to increased tissue hypoxia and elevated hypoxia inducible factor 1-*α* (HIF1-*α*) expression, which was suggested to contribute the inflammatory cascade (Lumeng et al., 2007; Palmer and Clegg, 2014; Wensveen et al., 2015). Since adiposity patterns, insulin sensitivity, LPS levels, TLR activation, hypoxia, and AT inflammation were all ameliorated in the presence of estrogen, its enhancement of AT metabolic state via GM modulation can be one of the protective roles female sex hormones possess against cardiometabolic dysfunction. Interestingly, HFD-fed androgen receptor knockout male mice had increased obesity, visceral adiposity and adipocyte hypertrophy, glucose intolerance, and insulin resistance compared with control males and HFD-fed females. These derangements were linked to dysbiosis as antibiotic treatment corrected the dysfunction (Harada et al., 2020). These observations further emphasize the complexity and the importance of sex hormone contribution to the GM-cardiometabolic interaction, particularly the female sex hormones playing a modulatory role on several intermediary factors of cardiometabolic insults as outlined above. Figure 2 depicts the different pathways triggered by GM, leading to WAT inflammation as well as their modulation by sex hormones. ![Fig. 2.](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F3.medium.gif) [Fig. 2.](http://molpharm.aspetjournals.org/content/102/1/60/F3) Fig. 2. Detrimental effects of dysbiosis on adipose tissue homeostasis. Gut microbiota dysbiosis amplifies high-fat diet-mediated adipose tissue dysfunction through increasing energy absorption, weight gain, and adiposity. The subsequent development of insulin resistance and adipose tissue inflammation leads to the development of the metabolic syndrome linked to the emergence of cardiovascular diseases. Testosterone and estrogen inhibit TLR4 signaling. Estrogen decreases the level of circulating LPS, enhances leptin sensitivity, decreases insulin resistance, and limits consequences of hypoxia by induction of the proteasomal degradation of HIF-1*α*. Several therapeutic interventions such as antibiotic treatment and fecal microbiota transfer also positively modulate the depicted pathways. Pathways implicated in gut microbiota dysbiosis are presented in red, whereas those counteracting them are presented in black. AT; ATB, antibiotic; CVD; FMT; GM; HFD; HIF-1*α*, hypoxia-inducible factor 1*α*; IL; MyD88; NF-κB, nuclear factor κ B; TLR4; TNF-*α*. Nonetheless, it is prudent to mention that accumulating evidence recognize sex hormone independent differences in metabolism and response to metabolic challenge between males and females (Manwani et al., 2015). Differences in consequences of the genetic disparity between the XX and XY chromosome combination extend well beyond sex hormone production to comprise variable levels of X-linked gene imprinting and expression, in addition to a considerable dimorphism in the complement of noncoding RNA molecules production (Link et al., 2013). The study of the metabolic sequalae of such differences was hampered for a long time by the lack of ability of dissociating the concurrence of the XX or the XY combination with the corresponding gonadal hormone production. Evidence from transgenic mouse models expressing different combinations of sex chromosomes in presence and absence of the corresponding gonads showed a remarkable impact of the XX chromosome combination on preferential subcutaneous AT distribution regardless of the gonadal hormone status (Chen et al., 2012). However, the increased subcutaneous adiposity in this model was associated with increased insulin resistance, thus emphasizing the protective effect of estrogen in this regard. More recently, genetic association studies were conducted on humans to detect the effect of the X chromosome on metabolic and cardiovascular disease but had conflicting results. Although some studies found differential associations with insulin resistance, atherosclerosis, and coronary artery disease between sexes (Tukiainen et al., 2014; Traglia et al., 2017), others failed to detect these differences and attributed the observations to gonadal hormones (Manwani et al., 2015). However, to our knowledge, the interaction between sex chromosomes and dysbiosis did not receive much attention and warrants detailed investigation in the future. #### Sex-Dependent Differences in Alteration of Short-Chain Fatty Acids Production and Consequent Metabolic Dysfunction Another important component of HFD-induced dysbiosis is the decline in SCFA generation, mainly acetate, propionate, and butyrate, which were correlated with numerous metabolic and cardiovascular disorders (Canfora et al., 2015; Chambers et al., 2018). Despite the extensive research on the role of SCFA in the host wellbeing, the exact role of SCFAs in regulating metabolic function remains debatable. For instance, some evidence suggested a negative correlation of fecal SCFAs levels with the host’s metabolic function (Teixeira et al., 2013; de la Cuesta-Zuluaga et al., 2018) with studies showing that SCFA levels were higher As for acetate and propionate, it is suggested that they offset LPS induced endotoxemia by lowering TNF-*α* and NF-*κ*B production as shown in human neutrophils and macrophages *in vitro* experiments (Canfora et al., 2015). These two SCFAs were shown to be effective in reducing TNF*α* production in LPS-activated neutrophils, while repressing the activity of NF-*κ*B receptor in a human colon adenocarcinoma cell line (Tedelind et al., 2007). Consistently, in vitro treatment of human omental and subcutaneous adipocytes with propionate, reduced mRNA expression of the proinflammatory factor resistin, and stimulated leptin mRNA expression (Al‐Lahham et al., 2010). Another study on human omental AT supported the previous findings, as propionate treatment declined both mRNA and protein levels of proinflammatory cytokines like IL-4 and TNF-*α* (Al‐Lahham et al., 2012). Additionally, animal models reared on HFD and treated with propionate had an improved insulin sensitivity, glucose tolerance, thermogenesis, and mitochondrial function, besides an improved metabolic state in brown adipose tissue, liver, and muscles (Liang and Ward, 2006; Brial et al., 2018). In humans, rectal administration of sodium acetate reduced serum TNF-*α* in obese females (Freeland and Wolever, 2010). Another trial on acute intravenous infusion of acetate in women with hyperinsulinemia and overweight improved serum peptide YY, glucagon-like peptide 1 and reduced circulating TNF*α* and ghrelin (Freeland and Wolever, 2010). Pathways modulated by SCFAs are summarized in Figure 3. ![Fig. 3.](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F4.medium.gif) [Fig. 3.](http://molpharm.aspetjournals.org/content/102/1/60/F4) Fig. 3. Short-chain fatty acids regulate lipogenic, inflammatory, and neuronal pathways that are dysregulated in states of metabolic dysfunction. The three major short-chain fatty acids are acetate, propionate, and butyrate. They improve cardiometabolic health through several pathways as indicated, thus counteracting HFD-induced metabolic dysfunction. Akt; BP; FIAF, fasting-induced adipose factor; FMT; GPR; HFD; IL; LPL, lipoprotein lipase; LPS; MAPK, mitogen-activated protein kinase; NF-κB; PKC; protein kinase C; PLC; PTEN, phosphatase and tensin homolog; SCFA; SREBP-1; TNF-*α*; UCP-1, uncoupling protein 1. Importantly, recent literature reveals that interventions targeting GM have a differential impact on SCFA generation across sexes. For instance, ciprofloxacin-metronidazole treatment reduced SCFA production only in female mice consistent with a reduction in the relative abundance of *Firmicutes* (Gao et al., 2019). Alternatively, prebiotic supplementation was shown to increase fecal butyrate output only in male but not female rats (Shastri et al., 2015). A similar study in humans demonstrated a differential effect whereby beta-glucan supplementation led to an increased butyrate production in female subjects but not in males (Trimigno et al., 2017). On the other hand, SCFAs were proposed to promote the storage of triglycerides through the activation of lipogenic hepatic enzymes including SREBP, which show a positive differential expression in women (Bäckhed et al., 2004; Jiang et al., 2016). This is suggested to reflect in an increased microbiota-dependent lipid storage and obesity risk in women. SCFAs also are known to suppress the fasting-induced adipocyte factor, an inhibitor of lipoprotein lipase (Bäckhed et al., 2004; Khan et al., 2016). This increased lipoprotein lipase activity may lead to microbiota-dependent augmentation in fat storage, which may contribute in part to sex differences in body composition (Bäckhed et al., 2004). Henceforth, sex-differential response to GPR41 may contribute to microbiota-associated body weight sexual dimorphism (Inoue et al., 2014). This is particularly important as male but not female GPR41 knockout mice exhibited an increased body fat mass and a decreased energy expenditure (Bellahcene et al., 2013). Moreover, further sex-specific interactions with SCFAs were reported where butyrate was shown to increase estrogen secretion in granulosa cell culture models (Lu et al., 2017). Indeed, conclusions about the involvement of SCFAs in cardiometabolic derangements in sex-dependent manner are hard to be drawn, and further studies are highly needed for more solid evidence. ## Sex-Dependent Impact of Gut Microbiome on Metabolically Induced Cardiovascular Dysfunction Although traditional cardiovascular risk factors appear to be related to the development of cardiovascular disease in either sex, research has long recognized significant complexity in their differential roles and relative weights (Njølstad et al., 1996). For instance, analysis of a large case-control study showed that the impact of diabetes and hypertension was more pronounced on the development of myocardial infarction in women than in men (Anand et al., 2008). Yet, the impact of these two factors appeared to differ by age, being stronger in younger men, leading to an age difference of cardiovascular disease onset by about nine years. This further implicates the role of sex hormones in the observed protective effect in premenopausal females, as the incidence of both hypertension (Lima et al., 2012) and diabetes (Heianza et al., 2013) in postmenopausal females appear to exceed that in men. Interestingly, it is well recognized that both disorders have strong mechanistic links to adipose tissue dysfunction, particularly perivascular adipose tissue (PVAT), observed in metabolic impairment (Saxton et al., 2019). Significantly, female-specific risk factors for cardiovascular disease such as polycystic ovary syndrome and preeclampsia appear to have a strong metabolic impairment component carrying the hallmarks of a dysfunctional adipose tissue (Huda et al., 2017; Leon et al., 2019; Osibogun et al., 2020). In the below sections, we examined the impact of sex-based differences in the interconnection among dysbiosis, metabolic impairment, and adipose inflammation on incidence of cardiovascular disease. #### Dysbiosis and Cardiovascular Dysfunction HFD is known to induce cardiovascular dysfunction (Martins et al., 2015; Aghajani et al., 2017). Since HFD stimulates dysbiosis, the microbiome-cardiovascular axis was extensively studied, and dysbiosis was linked to several diseases such as hypertension (HTN), atherosclerosis, and heart failure (HF) among others (Tang and Hazen, 2017; Taylor and Takemiya, 2017; Kappel and Federici, 2019; Razavi et al., 2019). For instance, atherosclerotic plaques were found to contain bacterial DNA, and these bacterial taxa were also present in the gut of the same individuals (Ott et al., 2006; Koren et al., 2011), proposing a possible role of microbial communities in plaque instability and the subsequent adverse effects (Koren et al., 2011). In patients with HF, both metabolites and gut flora print were significantly determinantal compared with healthy subjects and were even worse in patients with decompensated heart failure (Hayashi et al., 2018). Significantly, gut dysfunction involving disturbances in intestinal motility and villi absorption, in addition to an impaired tissue perfusion and edema, was also observed in HF patients (Krack et al., 2005; Sandek et al., 2012). Undeniably, the HFD-induced impairment of gut integrity and gut hyperpermeability were linked to the aforementioned cardiovascular insults (Lewis and Taylor, 2020). Gut microbial signature in HTN was heavily investigated in the last few decades (Mell et al., 2015; Kim et al., 2018). Interestingly, fecal microbial transplantation from hypertensive patients to germ-free mice induced HTN in these mice (Li et al., 2017). Moreover, these results were also observed in germ-free rats receiving GM from spontaneously hypertensive rats (Shi et al., 2021). However, the impact of GM appears to be complex as germ-free rats demonstrated a reversal of poor vascular contractility and reduced blood pressure control upon acquisition of normal GM (Joe et al., 2020). Indeed, a decrease in microbial richness and diversity in prehypertensive and hypertensive human subjects were recorded as well (Li et al., 2017). Additionally, spontaneously hypertensive and chronic angiotensin-II-induced hypertensive rat models presented dysbiosis manifested by an increase in *F*/*B* ratio compared with the normotensive controls (Yang et al., 2015; Santisteban et al., 2017). Moreover, high-salt diet was shown to deplete a strain of *Lactobacillus*; however, treating these mice with this strain attenuated salt-sensitive hypertension (Wilck et al., 2017). Furthermore, HFD-induced dysbiosis was shown to have a role in the development of the obstructive sleep apnea-induced HTN (Durgan et al., 2016). HFD-induced dysbiosis was also correlated with increased serum LPS-binding protein, IL-6, endothelial dysfunction, arterial stiffness, aortic phosphorylated NF-*κ*B, and NADPH oxidase in PVAT, leading to a positive oxidative state. Interestingly, all these insults were attenuated with antibiotic treatment (Battson et al., 2018). Not only had been NADPH oxidase (NOX)-related reactive oxygen species shown to be detrimental in CVDs (Brandes et al., 2010), PVAT inflammatory and oxidative changes were consistently reported to contribute to vascular and cardiac autonomic dysfunction in HFD-fed rats even prior to the development of overt metabolic impairment (Al-Assi et al., 2018; Elkhatib et al., 2019; Rafeh et al., 2020), implicating the GM-metabolic-AT-CVD axis, which will be discussed later. SCFAs may also contribute to the reduction of systemic blood pressure and serum cholesterol levels (Den Besten et al., 2013; Mariño et al., 2017). High-fiber diet, diet supplemented with SCFA, or parenteral injection of SCFA improved cardiometabolic health in several murine models by reducing blood pressure and cardiac fibrosis (Brial et al., 2018). It was proposed that the role of SCFA in blood pressure (BP) regulation might be mediated by the activation of GPR41 in the vascular endothelium (Jonsson and Bäckhed, 2017). As well, SCFAs were found to hold strong vasorelaxant properties (Poll et al., 2020). Interestingly, they were also discovered to be ligands for olfactory receptor 78, a G protein-coupled receptor expressed in the vasculature, which plays an important role in vasoregulation and renin release and is activated mainly by acetate and propionate. Olfactory receptor 78 knockout mice had basal hypotension and low serum renin level, possibly indicative of the opposing response of GPR41 to SCFAs (Pluznick et al., 2013; Pluznick, 2014). Importantly, SCFAs may also participate in regulating the sympathetic tone. Actually, in GPR41 knockout male mice, propionate was found to be a potent activator of sympathetic ganglia through G*βγ*-PLC*β*-MAPK pathway rather than cAMP synthesis inhibition (Kimura et al., 2011). Interestingly, propionate was able to trigger epinephrine secretion in sympathetic neurons. On the other hand, wild-type mice did not have GPR41 expression in the mesenteric fat pad and only presented GPR43 whose activation seemed to increase leptin production. Henceforth, it was suggested that GPR43 mediate sympathetic stimulation by adipocytes activation and leptin overexpression (Kimura et al., 2011). A recent study on Wistar-Kyoto and spontaneously hypertensive male rats revealed the strong association between dysbiosis and sympathetic activation through the induction of inflammation and oxidative stress in the brain. Not only blood pressure was corrected after fecal microbial transplantation from Wistar-Kyoto to spontaneously hypertensive rats, but also inflammation and oxidative stress in the paraventricular nucleus were improved. However, fecal microbial transplantation in the opposite direction deteriorated the inflammatory, oxidative, and blood pressure state of the Wistar-Kyoto rats simultaneously with poor gut integrity and increased colonic TNF-*α* and circulating LPS, which were attenuated in the former transplant. Worth mentioning, spontaneously hypertensive rats had a lower butyrate receptor expression in the hypothalamus, alongside a higher Th17 cells and macrophage infiltration in the paraventricular nucleus (Toral et al., 2019). Moreover, LPS infusion was found to induce HTN in normotensive rats, by provoking neuroinflammation in the rostral ventrolateral medulla, which is considered an important part in intensifying the sympathetic stream to the blood vessels (Wu et al., 2012). Interestingly, another study on the same model indicated a similar pattern of dysbiosis; increased *F*/*B* ratio, in addition to a notable reduction in acetate, increased gut sympathetic outflow, elevated blood pressure, impaired gut integrity, impaired endothelial-dependent relaxation to acetylcholine, and NOX overactivation. Treating spontaneously hypertensive rats with losartan, an angiotensin 2 receptor (AT2-R) antagonist, preserved gut integrity and enhanced functionality and immune response in the vasculature, improving acetylcholine-induced relaxation and increased Tregs infiltration (Santisteban et al., 2017; Robles‐Vera et al., 2020). Indeed, these lines of evidence support the involvement of dysbiosis in inducing diet-related HTN and sympathetic overactivation, alongside with neuroinflammation and oxidation, which is proposed to be corrected by SCFAs. Figure 4 depicts the role of GM in the interaction between the dysbiosis, cardiometabolic dysfunction, and neuroinflammation. ![Fig. 4.](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F5.medium.gif) [Fig. 4.](http://molpharm.aspetjournals.org/content/102/1/60/F5) Fig. 4. Gut microbiota dysbiosis in the metabolic dysfunction-neuroinflammation-cardiovascular disease continuum. Gut microbiota increases the *F*/*B* ratio resulting in an oxidative stress in the brain leading to neuroinflammation and sympathetic overactivation. The latter consequently increases blood pressure and predisposes to the development of hypertension. Gut microbiota dysbiosis also leads to endotoxemia and increases Th17 and macrophage brain infiltration, both leading to arterial hypertension. Gut microbiota dysbiosis also detrimentally accelerates adipose tissue dysfunction, leading to an increased production of leptin, which further augments the activation of the sympathetic system. Additionally, endotoxemia inhibits the rather beneficial accumulation of Treg cells in the brain, which further augments inflammation. Estrogen inhibits the development of hypertension through an eNOS-mediated vasodilatory effect and through decreasing serum levels of AngII. Progesterone as well as fecal microbiota transfer inhibits neuroinflammation and its downstream consequences. The antagonism of AT2-R also reverses immune cell profile alterations mediated by gut microbiota dysbiosis. Pathways involved in gut microbiota dysbiosis are presented in red, whereas those counteracting them in black. AngII, angiotensin II; AT; AT2-R; BP; eNOS, endothelial nitric oxide synthase;* F*/*B*; FMT; GM; LPS; Th17, T helper cell 17; Treg. On another note, accumulating evidence highlighted the role of dysbiosis in augmenting the production of the bacterial metabolite trimethylamine-N-oxide (TMAO), which is suggested to be an indicator of CVDs (Moludi et al., 2020). TMAO is a plasma metabolite formed through a metaorganismal pathway, and its level is depending on dietary intake mainly from animal protein sources like red meat, egg yolk, and seafood, which are abundant in choline, phosphatidylcholine, and L-carnitine (Anders et al., 2013; Tang et al., 2015). These compounds are initially metabolized by GM to form trimethylamine (TMA) and then converted by the host liver enzyme flavinmonooxygenase 3 (FMO3) to form TMAO (Bennett et al., 2013). Noteworthy, FMO3 knockdown in female mice protected from diet-induced obesity and reduced hypertrophy and adiposity in WAT while improving total metabolic health (Schugar et al., 2017). Increased TMAO has been anticipated to induce insulin resistance and adipose inflammation in mice as well as increasing the risk for type 2 diabetes in human subjects (Tang et al., 2017). Circulating TMAO was linked to elevated vascular inflammation through the incitement of proinflammatory cytokines expression and leukocytes recruitment (Seldin et al., 2016). Also, dietary supplementation of choline in mice increased TMAO levels, macrophage foam cell formation, and atherosclerosis incidence (Wang et al., 2011). Moreover, it prompted platelet hyperactivity (Trip et al., 1990; Marcucci et al., 2014) and enhanced thrombosis (Zhu et al., 2016), which are considered major risk factors for developing CVDs. Indeed, serum TMAO level was tightly correlated with atherosclerosis (Tang and Hazen, 2017). In the same context, a human study revealed that subjects with higher serum TMAO levels had a twofold risk increase for developing major cardiovascular events compared with subjects with low TMAO (Tang et al., 2013). TMAO levels had also been suggested to be an accurate indicator for heart failure diagnosis (Tang et al., 2014). It was found to fuel endothelial dysfunction as well by upregulating vascular adhesion molecule-1, monocyte attraction, and NF-*κ*B activation (Ma et al., 2017). As stated before, HFD induces AT dysfunction, which predispose to metabolic and cardiovascular disorders. The AT surrounding the vascular bed, referred to as PVAT, which has been identified as a crucial component of the vascular regulatory machinery. It is worth mentioning that PVAT has been suggested to be one of the most sensitive AT depots to positive energy intake and the first to undergo negative remodeling including hypertrophy, inflammation, and hypoxia, which were observed in early metabolic impairment (Elkhatib et al., 2019; AlZaim et al., 2020). Moreover, resistance arterioles from obese mice showed a PVAT-dependent impairment in insulin/Akt-mediated vasodilatation due to reduced adiponectin and AMPK downstream effects, which was restored with Jun NH2-terminal kinase inhibition (Meijer et al., 2013). HFD induced PVAT dysfunction might be mediated via sympathetic overactivation and insulin resistance that have been linked to a wide range of subclinical cardiovascular insults such as endothelial dysfunction and cardiac autonomic neuropathy (Britton and Fox, 2011; Bulloch and Daly, 2014; Greenstein et al., 2009; Akoumianakis et al., 2017; Alaaeddine et al., 2019; AlZaim et al., 2020; Bakkar et al., 2020; Rafeh et al., 2020). As such, it becomes plausible that PVAT inflammation might mediate the effect of HFD-induced dysbiosis on early metabolic impairment and cardiovascular dysfunction. This hypothesis is presented in Figure. 5. However, limited studies have explored the association between PVAT and HFD-induced dysbiosis. One recent study examined the role of PVAT FMO3 in response to direct TMA stimulation in tone regulation in excised aortas from male rats. TMA exerted a contractile effect through activating L-type voltage-gated calcium channels that was found to be dependent on endothelium rather than PVAT, suggesting that TMAO and TMA modulate vascular tone by a direct effect of vascular smooth muscle cells (Restini et al., 2021). However, to our knowledge no studies were conducted to investigate the impact of dysbiosis or SCFAs on PVAT modulation and thus the HFD-dysbiosis-PVAT axis remains elusive. ![Fig. 5.](http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/content/molpharm/102/1/60/F6.medium.gif) [Fig. 5.](http://molpharm.aspetjournals.org/content/102/1/60/F6) Fig. 5. Perivascular adipose tissue dysfunction: Novel mechanisms of gut microbiota dysbiosis-mediated cardiovascular derangements. A healthy perivascular adipose tissue secrets adiponectin, which elicits an AMPK-mediated anticontractile effect. The consumption of a high-fat diet causes sympathetic overactivation and insulin resistance, leading to perivascular adipose tissue expansion, adipocyte hypertrophy, and adipose tissue inflammation, thus jeopardizing the anticontractile activity of perivascular adipose tissue. Importantly, the consumption of a high-fat diet causes gut microbiota dysbiosis, which enhances NOX-mediated production of ROS, a pathway that is augmented after ovariectomy. Additionally, gut microbiota dysbiosis increases aortic NF-κB signaling, leading to arterial stiffness. Microbial metabolism products including TMA and TMAO participate in gut microbiota dysbiosis-caused perivascular adipose tissue dysfunction. TMA, through its activity on VSMCs induces L-type voltage-gated calcium channels, which counteracts perivascular adipose tissue-mediated anti-contractile effect. AMPK; GM; HFD; NOX; PVAT; ROS; TMA; TMAO; VCAM-1; VSMCs, vascular smooth muscle cells. #### Sex-Dependent Impact of Dysbiosis on Cardiovascular Dysfunction Sex-dependent cardiovascular risk and pathology are well documented in the literature, as men have a higher absolute risk compared with premenopausal women, a difference that diminishes after menopause, indicating the important role of sex hormones in CVDs (Kim and Reaven, 2013; Pei et al., 2017; WHO, 2017; Chella Krishnan et al., 2018). Although metabolic disorders and cardiovascular diseases have long been intertwined, early metabolic impairment has been the focus of interest as it imparts predisposition to inevitable CVDs. This effect is mainly mediated by subclinical events such as metabolic endotoxemia, AT inflammation, and insulin resistance (Heilbronn and Campbell, 2008; Shah et al., 2008; Nishimura et al., 2009; Kallio et al., 2015; Wensveen et al., 2015). Interestingly, GM has been identified as a major driver of these anomalies. Since sex-dependent differences exist in dysbiosis, the differential GM effect is expected to be extrapolated to CVDs. As discussed previously, estrogens attenuate HFD-induced gut hyperpermeability and LPS transport either through leaky membranes or chylomicrons together with the consequent metabolic endotoxemia, Th17 cell activation, and Tregs inhibition (Cani et al., 2007), which were found to be higher in men. As stated previously, LPS activates TLR4 on target tissues including AT and macrophages, triggering proinflammatory cascade and activating NF-*κ*B. AT overactivation and hypertrophied expansion due to dysbiosis was linked to increased leptin production, which in turn will activate sympathetic outflow. In parallel, LPS-induced neuroinflammation triggers sympathetic firing. The resultant sympathetic overactivation and insulin resistance, which are expected to be higher in males, will lead to early inflammation and negative remodeling of PVAT, precipitating a wide range of subclinical cardiovascular insults (Greenstein et al., 2009; Britton and Fox, 2011; Bulloch and Daly, 2014; Akoumianakis et al., 2017; Khatib et al., 2018; Alaaeddine et al., 2019; Bakkar et al., 2020). Since estrogen holds a protective effect against hypoxia in AT, possibly estrogen will block dysbiosis-mediated dysfunction cardiovascular dysfunction by interfering with PVAT inflammation. Additionally, progesterone was found to have an anti-inflammatory effect against LPS mediated neuroinflammation (Lei et al., 2014). The evidence regarding sexual dimorphism in HFD-induced PVAT remodeling is scarce; however, females tend to have a more functional PVAT compared with males. In this regard, ovariectomy in murine models instigated endothelial and PVAT dysfunction mediated by increased reactive oxygen species when compared with their sham littermate (Wang et al., 2014; Taylor and Sullivan, 2016). Additionally, an enhanced anticontractile role of PVAT was observed in female pigs and was attributed to a higher sensitivity of adiponectin receptor in coronary artery (Ahmad et al., 2017). Taken together, these observations suggest a possible role of PVAT in mediating a sex-dependent cardiovascular impact of dysbiosis in early metabolic dysfunction. On another note, the protective effect of estrogen on HTN development is mainly through inducing endothelial nitric oxide synthase mediated vasorelaxation (Sobrino et al., 2017; Bucci et al., 2002), while at the same time inhibiting vasoconstricting agents such as angiotensin-II (Schunkert et al., 1997). However, mounting evidence suggests the role of immune responses in mediating sex-dependent GM-HTN axis. For instance, dysbiosis was linked to an increased activity of Th17 cells, which had a role in initiating arterial hypertension (Guzik et al., 2007; Ivanov et al., 2009; Wenzel et al., 2016). In this regard, hypertensive male rats had higher Th17 activity compared with females (Gillis and Sullivan, 2016). ## Therapeutic Interventions for Cardiometabolic Consequences of Dysbiosis Bidirectional interactions between GM and cardiovascular drugs have been reported for quite some time. Indeed, not only has the gut bacterial community been implicated in altering the pharmacokinetics of some cardiovascular drugs, but treatment with certain drug classes has also been associated with favorable changes in GM populations. For instance, certain phyla of gut bacteria were shown to metabolize digoxin and amlodipine, reducing their availability at target tissues, whereas others were proposed to decrease the absorption of simvastatin and captopril (Tuteja and Ferguson, 2019). In parallel, one human study showed that the low-density lipoprotein cholesterol lowering effect of a 4–8-week rosuvastatin treatment was associated with a change in the abundance of *Firmicutes* (Liu et al., 2018). As for animal studies, atorvastatin therapy appeared to reverse HFD-induced dysbiosis in male rats (Khan et al., 2018). Similarly, captopril treatment reduced dysbiosis and improved gut permeability associated with hypertension in spontaneously hypertensive rats (Santisteban et al., 2017). However, none of these interventions has been examined systematically, and the underlying mechanisms remain unclear. From a different perspective, tailored pharmacological interventions targeting GM with the purpose of imparting protective cardiovascular research have been proposed. Indeed, as bacterial metabolic reactions have been thoroughly recognized, selective approaches could be designed to modify harmful metabolite production. As such, small molecule inhibitors of TMAO synthesis were designed and proposed to treat atherosclerosis (Wang et al., 2015). Moreover, nanoparticle-based approaches were proposed either to deliver useful bacterial species associated with increased SCFA production or reduced LPS, or to scavenge TMAO and proinflammatory cytokines (Kazemian et al., 2020). Nevertheless, all these interventions remain in early stages, and the current viable options for prevention of detrimental outcomes of gut bacterial alteration remain related to direct manipulation of bacterial population using probiotics, antibiotics, fecal microbial transplantation, or using bacterial metabolites such as SCFAs as described below. The impact of these interventions on dysbiosis triggered pathways are demonstrated throughout Figures 1–5. #### Probiotics Probiotics are nonpathogenic strains of bacteria, usually belonging to *Lactobacilli* and *Bifidobacteria,* which have been used to reset microbiome dysbiosis (Holzapfel and Schillinger, 2002; Isolauri et al., 2004; Williams, 2010). Some clinical trials attempted to explore probiotics as a potential intervention with the symptoms in some neurologic and psychologic diseases such as amiotrophic lateral sclerosis and schizophrenia (Severance et al., 2017; Mazzini et al., 2018), whereas others focused on using different strains of probiotic bacteria as a potential therapy and early preventive technique for cardiovascular and metabolic disorders. However, these trials have had controversial findings. On one hand, several studies showed that probiotic administration improved cardiometabolic and inflammatory parameters not only in metabolically impaired but also in borderline individuals. For instance, a double-blind placebo-controlled trial showed that daily ingestion of *Lactobacillus plantarum* in hypercholesteremic individuals for 12 weeks significantly improved blood pressure, reduced serum total cholesterol, LDL, and triglycerides, while increasing high-density lipoprotein (HDL) levels (Costabile et al., 2017). The same strain was used for 6 weeks in subjects who smoked and had similar findings, reducing CV risk factors (Naruszewicz et al., 2002). In postmenopausal women with metabolic syndrome, supplementation with the same strain for 90 days decreased blood glucose and homocysteine (Barreto et al., 2014). On the same note, *Lactobacillus plantarum* 299v supplementation for 6 weeks in men with stable coronary artery disease significantly improved endothelium dependent vasodilatation, induced some changes in GM by enriching *Lactobacillus* genus, decreased plasma propionate, leptin and IL-8 and 12, without changing blood glucose, lipid profile, and body weight (Malik et al., 2018). Another strain of bacteria, *Bifidobacterium longum* BB536, exhibited beneficial effects after 12 weeks of blinded controlled intervention of food supplement intake containing red yeast extract, niacin, and coenzyme Q10 on individuals with a low score of CV risk. The results showed in improved levels of atherogenic lipid profile (Ruscica et al., 2019). Men with mild hypercholesteremia were treated with isoflavone-supplemented soy product fermented with *Enterococcus faecium* CRL 183 and *Lactobacillus helveticus* 416 for 42 days and showed an improved serum lipid profile, but neither CRP nor fibrinogen (Cavallini et al., 2016). Another study examined the role of *Bifidobacterium lactis* in patients with metabolic syndrome. The organism was supplemented in fermented milk and given for 45 days. Treated subjects showed a reduction in BMI, serum TNF-*α*, and IL-6, while improving lipid profile (Bernini et al., 2016). A 2-month treatment with yogurt supplemented with a probiotic mix (*Lactobacillus acidophilus* La5 and *Bifidobacterium lactis* Bb12) in men and women with metabolic syndrome improved fasting blood glucose and insulin sensitivity. Importantly, it improved some vascular and endothelial function markers, like vascular cell adhesion molecule 1 (VCAM-1) and plasminogen activator inhibitor 1 (Rezazadeh et al., 2019). Healthy subjects with BMI at the upper limit of the healthy range randomized for a 12-week treatment with *Bifidobacterium lactis* with arginine supplementation appeared to have a better endothelial function and hence a reduced risk of developing atherosclerosis (Matsumoto et al., 2019). This suggests a possible therapeutic and preventive effect of probiotics on endothelial function and CV risk. However, conclusions should be drawn carefully from these results given the possible contribution of arginine supplementation to endothelial improvement and the intervention being on low-risk subjects. Interestingly and worth investigating is the interplay between sex hormones and probiotics, as these positive outcomes seem to be independent of the status of sex hormones. In obese postmenopausal women, administration of multispecies probiotic for 12 weeks seemed to improve metabolic parameters: serum insulin, glucose, LPS, total lipid profile, uric acid, and HOMA-IR in both high and low dose groups; however, reduced adiposity was only observed in the high-dose arm, suggesting an improved gut permeability and reduced cardiometabolic risk factors (Szulińska et al., 2018). Similarly, premenopausal women diagnosed with polycystic ovarian syndrome were treated with pomegranate juice with and without probiotics mix (*Lactobacillus rhamnosus* GG, *bacillus koagolans*, and* indicous*) for 8 weeks. The group receiving probiotics showed an improved metabolic and inflammatory function alongside reduction in blood pressure (Esmaeilinezhad et al., 2020). Women aged 20–50 years with arterial HTN treated with a probiotic cocktail (*Lactobacillus para casei* LPC-37, *Lactobacillus rhamnosus* HN001, *Lactobacillus acidophilus* NCFM, and *Bifidobacterium lactis* HN019) for 8 weeks showed an improved fasting blood glucose, cholesterol, and elevated HDL level compared with the baseline. Interestingly, probiotics improved autonomic function and heart rate variability by reducing the low frequency domain, without significantly changing blood pressure; yet systolic BP was reduced by 5 mmHg compared with the placebo (Romão da Silva et al., 2020) On the other hand, several clinical trials failed to record therapeutic benefits of probiotics on metabolic and cardiovascular outcomes. For instance, the commercial probiotic VSL#3,, which contains 8 different strains of lactic acid bacteria, was used in a twice daily intervention for 10 weeks in men and women with nonalcoholic fatty liver disease. It did not appear to improve cardiovascular risk factors and liver injury scores. Nevertheless, it improved HOMA-IR (Chong et al., 2021). Another randomized controlled crossover study showed that metabolic syndrome symptoms were not alleviated by a daily intervention with *Lactobacillus reuteri* V3401 strain for 12 weeks. Yet, this intervention was able to reduce some inflammatory markers, IL-6 and VCAM-1 (Tenorio-Jiménez et al., 2019). Although probiotics exhibited some beneficial metabolic outcomes in human trials, results seemed to be dependent on the bacterial species used. As such, some probiotics did not seem to change dysbiosis-related parameters such as gut permeability compared with the control groups (Leber et al., 2012; Ivey et al., 2015; Stadlbauer et al., 2015; Grąt et al., 2017). For instance, using *Lactobacillus casei* Shirota for 12 weeks in subjects with metabolic syndrome did not correct dysbiosis nor improve gut integrity (Stadlbauer et al., 2015). Thus, conclusions drawn from probiotics intervention must be specified to the strains and concentrations used. Another important note is that not only different strains have been used in probiotics studies, but even some trials used different approaches in implementing the interventions, such as using probiotics with other dietary components (Cavallini et al., 2016; Scorletti et al., 2018; Rezazadeh et al., 2019; Ruscica et al., 2019), or other dietary interventions and lifestyle modification,s such as calorie restriction and physical activity (Behrouz et al., 2017). Therefore, the role of probiotics in combating metabolic and cardiovascular insults must be carefully investigated, and studies should be accurately designed to limit other confounding factors such as dietary and lifestyle modifications. However, controlled use of probiotics can be safe and useful in preventing CVDs and metabolic derangements in low-risk individuals alone or in combination with other compounds such as prebiotics (Behrouz et al., 2017; Trotter et al., 2020). #### Antibiotics Antibiotics were proposed to be one of the interventions to achieve eubiosis (Ianiro et al., 2016). However, few studies explored the efficacy of antibiotics in ameliorating dysbiosis related cardiometabolic dysfunction. For example, one study used antibiotics to reset GM community in patients with type 2 diabetes mellitus and obesity. Yet, major metabolic parameters such as insulin sensitivity, systemic inflammation, gut permeability, and adipocyte size did not change positively in response to a 7-day treatment of amoxicillin, vancomycin, or placebo (Reijnders et al., 2016). One case study on a postmenopausal woman suffering from chronic resistant HTN for 3 years, which was uncontrolled on more than 3 antihypertensive drugs, in addition to a history of metabolic and immune pathologies including diabetes and arthritis, reported a temporary (6-month) improvement of her HTN control upon treatment with a postoperative antibiotic mix (IV vancomycin, rifampin, and ciprofloxacin orally) (Qi et al., 2015). Given the adverse effects associated with antibiotics use and the risk of development of antibacterial resistance, this might be the least desirable intervention to correct dysbiosis and related pathologies. #### Fecal Microbial Transplantation Fecal microbial transplantation (FMT) is a novel method that has been recently suggested to induce eubiosis and alleviate pathologies mediated by disturbed gut microbiome. It is the process of isolating GM from healthy donors to transplant it into diseased subjects. FMT can be done through various methods that are relatively safe and noninvasive, rectally like enema, naso-gastric route, or orally by capsules (Lagier, 2014; Wang et al., 2016). Transplanted microbiota can be homologous from the same person and heterologous/allogenic from first-degree relatives or other healthy subjects. Interestingly, allogenic GM transplantation was found to be more effective than homologous interventions (Grehan et al., 2010; Wang et al., 2016; Schepici et al., 2019). Adverse effects reported after FMT are not serious. The side effects reported were mainly abdominal discomfort and diarrhea for a few hours after the procedure (Lagier, 2014). However, there remains concerns about the potential safety/side effects related to the nonbacterial component of the fecal material transferred (Bojanova and Bordenstein, 2016). A preliminary report observed that sterile protein isolates from donor fecal material were able to induce the required response in recipients (Ott et al., 2017); however, future investigation will be required to determine the possibility of fractionation and reducing the content being transplanted to the necessary organisms only. FMT has been considered as one of the important lines of life-saving treatments for patients with *Clostridium difficile* infections as these patients had improved outcomes and less chances for reoccurrence than those receiving conventional treatments (Van Nood et al., 2013; Kelly et al., 2016; Lee et al., 2016). It was even suggested that FMT can be promising in eradicating multidrug resistant microorganisms (Saha et al., 2019). Since GM has been identified as an important variable in the pathogenicity and prognosis of a large set of metabolic and cardiovascular diseases, FMT might be effective in correcting and alleviating dysbiosis related dysfunctions, especially the ones starting early on and having no clear treatment regimens. One clinical trial on male subjects with metabolic derangements including hyperinsulinemia, BMI above 30, elevated waist circumference, and increased adiposity treated with purified GM from lean and healthy donors (allogenic transfer) matched in sex and age through a duodenal tube over a 6-week period showed improved insulin sensitivity and microbial diversity favoring butyrate producing bacteria such as *Roseburia intestinalis* compared with the control group receiving an autologous transfer (Vrieze et al., 2012). However, another double-blinded randomized clinical trial using FMT delivered through capsules once per week from healthy donors to subjects with obesity and insulin resistance did not record any difference in either parameter after 12 weeks of intervention (Reijnders et al., 2016). Similar results were recorded from a randomized trial of obese adolescents, after ingestion of 28 capsules of lean donors’ GM. Up to 26 weeks postintervention, recipients of either sex did not show any evidence of improvement neither in metabolic parameters such as insulin sensitivity nor in obesity, although the central to peripheral fat ratio was reduced in the FMT arm only (Leong et al., 2020). Furthermore, FMT in patients with nonalcoholic fatty liver disease who suffered from insulin resistance delivered directly to the colon from autologous and allogenic sources did not improve insulin resistance; however, allogenic FMT improved gut permeability (Craven et al., 2020). Also, FMT in patients with metabolic syndrome from a healthy vegan donor did not alleviate TMAO levels and vascular inflammation (Smits et al., 2018). Although FMT has been revolutionary in treating diseases like *Clostridium difficile* infections, ulcerative colitis, and others, its role in cardiometabolic dysfunction is not fully understood, requires further investigation, and its long-term efficacy has yet to be established (Zhang et al., 2019b). #### Short-Chain Fatty Acids In human studies, SCFAs were measured as a secondary outcome of dietary intervention rather than being the treatment per se, and most evidence is drawn from animal studies. One recent study using rectal capsule delivery of SCFAs in a triple-blinded randomized trial examined the effect of 1 week of SCFAs administration on psychosocial stress of 66 healthy men. In the two intervention arms, low and high SCFAs were equally successful in reducing cortisol levels in response to psychosocial stress, and both had an increased serum SCFAs as well compared with the placebo arm (Dalile et al., 2020). The OmniHeart study, which included 164 adults, assessed the role of macronutrients on serum SCFAs levels. Three isocaloric and high-fiber diets rich in either carbohydrate, protein, or unsaturated fat were applied for 6 weeks. The results indicated differences in SCFAs serum levels in response to different diets, which were correlated with some cardiometabolic aspects. For instance, the butyrate level was only increased by a high-protein diet and was associated with decreased HDL levels and ghrelin and increased insulin and glucose levels (Mueller et al., 2020). A lot of questions and concerns arise regarding the use of SCFAs for therapeutic purposes in humans, especially in cardio and metabolic pathologies. Specifically, therapeutic dose selection, safety and efficacy of single or combined use, and most importantly the long-term effects of their use. Henceforth, more clinical studies should be done using SCFAs as a therapy for early metabolic derangements in a sex-dependent fashion. ## Conclusion Dysbiosis is a common occurrence in patients suffering from cardiometabolic conditions. Not only do GM alterations in these patients appear to be driven by the same risk factors of the other pathologies, but they also seem to contribute to and drive the molecular changes leading to cardiovascular involvement, including AT inflammation, particularly in PVAT. Sexual dimorphism is evident is several steps starting at the differential effect of sex hormones on GM diversity and stability, encompassing sex-dependent effects on GM metabolite production, gut permeability, vulnerability of AT to inflammatory changes, and culminating in a different susceptibility to CVD incidence. Future investigation utilizing systematic approaches is required for a better understanding of the pathways involved to allow for tailored therapy for effective management of early cardiometabolic dysfunction in either sex. ## Authorship Contributions *Wrote or contributed to the writing of the manuscript:* Dwaib, AlZaim, Ajouz, Eid, El-Yazbi. ## Footnotes * Received June 14, 2021. * Accepted October 23, 2021. * This work was supported by an American University of Beirut (AUB) Faculty of Medicine MPP grant to A.E.-Y. H.S.D. is supported by a Ph.D. Scholarship from the Faculty of Agriculture and Food Sciences at AUB and a l’Oreal–UNESCO for Women in Science Fellowship. I.A. is supported by a MasterCard Foundation Scholarship. * No author has an actual or perceived conflict of interest with the contents of this article. * [https://doi.org/10.1124/molpharm.121.000338](https://doi.org/10.1124/molpharm.121.000338). ## Abbreviations Akt : protein kinase B AMPK : AMP-activated protein kinase AT : adipose tissue AT2-R : angiotensin 2 receptor BMI : body mass index BP : blood pressure CVD : cardiovascular disease E2 : 17*β*-estradiol ER*β* : estrogen receptor-*β* F/B : Firmicutes/Bacteroidetes FMO3 : flavinmonooxygenase 3 FMT : fecal microbial transplantation GM : gut microbiota GPR : G-protein coupled receptor HDL : high-density lipoprotein HF : heart failure HFD : high-fat diet HOMA-IR : homeostatic model assessment–insulin resistance HTN : hypertension IL : interleukin LPS : lipopolysaccharide MUC2 : mucin 2 MyD88 : myeloid differentiation primary response 88 NF-*κ*B : nuclear factor kappa-light-chain-enhancer of activated B cells NOX : NADPH oxidase PGC-1*α* : PPAR*γ* gamma coactivator-1*α* PLC : phospholipase C PVAT : perivascular adipose tissue SCFA : short chain fatty acids SREBP-1 : sterol response element binding protein-1 TLR4 : toll-like receptor 4 TMA : trimethylamine TMAO : trimethylamine-N-oxide TNF*α* : tumor necrosis factor *α* Tregs : regulatory T cells VCAM-1 : vascular cell adhesion protein 1 WAT : white adipose tissue * Copyright © 2022 by The American Society for Pharmacology and Experimental Therapeutics ## References 1. Abdul-Aziz MA, Cooper A, and Weyrich LS (2016) Exploring relationships between host genome and microbiome: new insights from genome-wide association studies. Front Microbiol 7:1611. 2. Acharya KD, Gao X, Bless EP, Chen J, and Tetel MJ (2019) Estradiol and high fat diet associate with changes in gut microbiota in female ob/ob mice. Sci Rep 9:20192. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/s41598-019-56723-1&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=31882890&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 3. Aghajani M, Imani A, Faghihi M, Vaez Mahdavi MR, Mahboubi S, Moradi F, and Kazemi Moghaddam E (2017) Does increased nitric oxide production and oxidative stress due to high fat diet affect cardiac function after myocardial infarction? J Cell Mol Anesth 2:3–8. 4. Ahmadmehrabi S and Tang WHW(2017) Gut microbiome and its role in cardiovascular diseases. Curr Opin Cardiol 32:761–766. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/HCO.0000000000000445&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=29023288&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 5. Ahmad AA, Randall MD, and Roberts RE (2017) Sex differences in the regulation of porcine coronary artery tone by perivascular adipose tissue: a role of adiponectin? Br J Pharmacol 174:2773–2783. 6. Akoumianakis I, Tarun A, and Antoniades C (2017) Perivascular adipose tissue as a regulator of vascular disease pathogenesis: identifying novel therapeutic targets. Br J Pharmacol 174:3411–3424. 7. Alaaeddine RA, Mroueh A, Gust S, Eid AH, Plane F, and El-Yazbi AF (2019) Impaired cross-talk between NO and hyperpolarization in myoendothelial feedback: a novel therapeutic target in early endothelial dysfunction of metabolic disease. Curr Opin Pharmacol 45:33–41. 8. Al-Assi O, Ghali R, Mroueh A, Kaplan A, Mougharbil N, Eid AH, Zouein FA, and El-Yazbi AF (2018) Cardiac autonomic neuropathy as a result of mild hypercaloric challenge in absence of signs of diabetes: modulation by antidiabetic drugs. Oxid Med Cell Longev 2018:9389784. 9. Alderman MH (2021) Prediabetes: an unexplored cardiovascular disease risk factor. J Hypertens 39:42–43. 10. Al-Lahham SH, Roelofsen H, Priebe M, Weening D, Dijkstra M, Hoek A, Rezaee F, Venema K, and Vonk RJ (2010) Regulation of adipokine production in human adipose tissue by propionic acid. Eur J Clin Invest 40:401–407. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1111/j.1365-2362.2010.02278.x&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20353437&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000276924300004&link_type=ISI) 11. Al-Lahham S, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, and Venema K (2012) Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest 42:357–364. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1111/j.1365-2362.2011.02590.x&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=21913915&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 12. Allin KH, Tremaroli V, Caesar R, Jensen BAH, Damgaard MTF, Bahl MI, Licht TR, Hansen TH, Nielsen T, Dantoft TM, et al; IMI-DIRECT consortium (2018) Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia 61:810–820. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s00125-018-4550-1&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 13. AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, and El-Yazbi AF (2020) Adipose tissue immunomodulation: a novel therapeutic approach in cardiovascular and metabolic diseases. Front Cardiovasc Med 7:602088. 14. Anand SS, Islam S, Rosengren A, Franzosi MG, Steyn K, Yusufali AH, Keltai M, Diaz R, Rangarajan S, and Yusuf S; INTERHEART Investigators (2008) Risk factors for myocardial infarction in women and men: insights from the INTERHEART study. Eur Heart J 29:932–940. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1093/eurheartj/ehn018&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18334475&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000254684900019&link_type=ISI) 15. Anders HJ, Andersen K, and Stecher B (2013) The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 83:1010–1016. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/ki.2012.440&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23325079&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000319740300010&link_type=ISI) 16. Ang Z and Ding JL (2016) GPR41 and GPR43 in obesity and inflammation—protective or causative? Front Immunol 7:28. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.3389/fimmu.2016.00028&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26870043&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 17. Ash-Bernal R and Peterson LR (2006) The cardiometabolic syndrome and cardiovascular disease. J Cardiometab Syndr 1:25–28. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17675903&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 18. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, and Gordon JI (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 101:15718–15723. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTAxLzQ0LzE1NzE4IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. Bäckhed F, Manchester JK, Semenkovich CF, and Gordon JI (2007) Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 104:979–984. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo5OiIxMDQvMy85NzkiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 20. Bakkar NZ, Dwaib HS, Fares S, Eid AH, Al-Dhaheri Y, and El-Yazbi AF (2020) Cardiac autonomic neuropathy: a progressive consequence of chronic low-grade inflammation in type 2 diabetes and related metabolic disorders. Int J Mol Sci 21:9005. 21. Barreto FM, Colado Simão AN, Morimoto HK, Batisti Lozovoy MA, Dichi I, and Helena da Silva Miglioranza L (2014) Beneficial effects of lactobacillus plantarum on glycemia and homocysteine levels in postmenopausal women with metabolic syndrome. Nutrition 30:939–942. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.nut.2013.12.004&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24613434&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 22. Battson ML, Lee DM, Jarrell DK, Hou S, Ecton KE, Weir TL, and Gentile CL (2018) Suppression of gut dysbiosis reverses Western diet-induced vascular dysfunction. Am J Physiol Endocrinol Metab 314:E468–E477. 23. Behrouz V, Jazayeri S, Aryaeian N, Zahedi MJ, and Hosseini F (2017) Effects of probiotic and prebiotic supplementation on leptin, adiponectin, and glycemic parameters in non-alcoholic fatty liver disease: a randomized clinical trial. Middle East J Dig Dis 9:150–157. 24. Bellahcene M, O’Dowd JF, Wargent ET, Zaibi MS, Hislop DC, Ngala RA, Smith DM, Cawthorne MA, Stocker CJ, and Arch JR (2013) Male mice that lack the G-protein-coupled receptor GPR41 have low energy expenditure and increased body fat content. Br J Nutr 109:1755–1764. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1017/S0007114512003923&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23110765&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 25. Bellenger J, Bellenger S, Escoula Q, Bidu C, and Narce M (2019) N-3 polyunsaturated fatty acids: an innovative strategy against obesity and related metabolic disorders, intestinal alteration and gut microbiota dysbiosis. Biochimie 159:66–71. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.biochi.2019.01.017&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 26. Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, Allayee H, Lee R, Graham M, Crooke R, et al. (2013) Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab 17:49–60. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cmet.2012.12.011&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23312283&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000313998800008&link_type=ISI) 27. Bernini LJ, Simão ANC, Alfieri DF, Lozovoy MAB, Mari NL, de Souza CHB, Dichi I, and Costa GN (2016) Beneficial effects of Bifidobacterium lactis on lipid profile and cytokines in patients with metabolic syndrome: a randomized trial. Effects of probiotics on metabolic syndrome. Nutrition 32:716–719. 28. Bless EP, Reddy T, Acharya KD, Beltz BS, and Tetel MJ (2014) Oestradiol and diet modulate energy homeostasis and hypothalamic neurogenesis in the adult female mouse. J Neuroendocrinol 26:805–816. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1111/jne.12206&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25182179&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 29. Boffa LC, Vidali G, Mann RS, and Allfrey VG (1978) Suppression of histone deacetylation in vivo and in vitro by sodium butyrate. J Biol Chem 253:3364–3366. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjExOiIyNTMvMTAvMzM2NCI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 30. Bojanova DP and Bordenstein SR (2016) Fecal transplants: what is being transferred? PLoS Biol 14:e1002503. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pbio.1002503&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27404502&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 31. Bouloumié A, Casteilla L, and Lafontan M (2008) Adipose tissue lymphocytes and macrophages in obesity and insulin resistance: makers or markers, and which comes first? Arterioscler Thromb Vasc Biol 28::1211–1213. [FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYXR2YmFoYSI7czo1OiJyZXNpZCI7czo5OiIyOC83LzEyMTEiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 32. Bouloumié A, Curat CA, Sengenès C, Lolmède K, Miranville A, and Busse R (2005) Role of macrophage tissue infiltration in metabolic diseases. Curr Opin Clin Nutr Metab Care 8:347–354. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/01.mco.0000172571.41149.52&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=15930956&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000232728200001&link_type=ISI) 33. Brandes RP, Weissmann N, and Schröder K (2010) NADPH oxidases in cardiovascular disease. Free Radic Biol Med 49:687–706. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.freeradbiomed.2010.04.030&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20444433&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000280745900001&link_type=ISI) 34. Braniste V, Leveque M, Buisson-Brenac C, Bueno L, Fioramonti J, and Houdeau E (2009) Oestradiol decreases colonic permeability through oestrogen receptor beta-mediated up-regulation of occludin and junctional adhesion molecule-A in epithelial cells. J Physiol 587:3317–3328. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1113/jphysiol.2009.169300&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19433574&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 35. Brial F, Le Lay A, Dumas M-E, and Gauguier D (2018) Implication of gut microbiota metabolites in cardiovascular and metabolic diseases. Cell Mol Life Sci 75:3977–3990. 36. Britton KA and Fox CS (2011) Perivascular adipose tissue and vascular disease. Clin Lipidol 6:79–91. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.2217/clp.10.89&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=21686058&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 37. Bryzgalova G, Lundholm L, Portwood N, Gustafsson J-A, Khan A, Efendic S, and Dahlman-Wright K (2008) Mechanisms of antidiabetogenic and body weight-lowering effects of estrogen in high-fat diet-fed mice. Am J Physiol Endocrinol Metab 295:E904–E912. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpendo.90248.2008&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18697913&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000259967600019&link_type=ISI) 38. Bucci M, Roviezzo F, Cicala C, Pinto A, and Cirino G (2002) 17-β-oestradiol-induced vasorelaxation in vitro is mediated by eNOS through hsp90 and akt/pkb dependent mechanism. Br J Pharmacol 135:1695–1700. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/sj.bjp.0704641&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=11934809&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000174940800013&link_type=ISI) 39. Bulloch JM and Daly CJ (2014) Autonomic nerves and perivascular fat: interactive mechanisms. Pharmacol Ther 143:61–73. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.pharmthera.2014.02.005&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24560685&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 40. Cândido TLN, da Silva LE, Tavares JF, Conti ACM, Rizzardo RAG, and Gonçalves Alfenas RC (2020) Effects of dietary fat quality on metabolic endotoxaemia: a systematic review. Br J Nutr 124:654–667. 41. Candon S, Perez-Arroyo A, Marquet C, Valette F, Foray A-P, Pelletier B, Milani C, Ventura M, Bach J-F, and Chatenoud L (2015) Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes. PLoS One 10:e0125448. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0125448&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25970503&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 42. Canfora EE, Jocken JW, and Blaak EE (2015) Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 11:577–591. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nrendo.2015.128&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26260141&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 43. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56:1761–1772. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6OToiNTYvNy8xNzYxIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 44. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, and Burcelin R (2008) Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57:1470–1481. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6OToiNTcvNi8xNDcwIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 45. Carmody RN, Gerber GK, Luevano JM Jr., Gatti DM, Somes L, Svenson KL, and Turnbaugh PJ (2015) Diet dominates host genotype in shaping the murine gut microbiota. Cell Host Microbe 17:72–84. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.chom.2014.11.010&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25532804&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 46. Cavallini DC, Manzoni MS, Bedani R, Roselino MN, Celiberto LS, Vendramini RC, de Valdez G, Abdalla DS, Pinto RA, Rosetto D, et al. (2016) Probiotic soy product supplemented with isoflavones improves the lipid profile of moderately hypercholesterolemic men: a randomized controlled trial. Nutrients 8:52. 47. Chambers ES, Preston T, Frost G, and Morrison DJ (2018) Role of gut microbiota-generated short-chain fatty acids in metabolic and cardiovascular health. Curr Nutr Rep 7:198–206. 48. Chang E, Varghese M, and Singer K (2018) Gender and sex differences in adipose tissue. Curr Diab Rep 18:69. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s11892-018-1031-3&link_type=DOI) 49. Chella Krishnan K, Mehrabian M, and Lusis AJ (2018) Sex differences in metabolism and cardiometabolic disorders. Curr Opin Lipidol 29:404–410. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/MOL.0000000000000536&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=30156571&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 50. Chen X, McClusky R, Chen J, Beaven SW, Tontonoz P, Arnold AP, and Reue K (2012) The number of x chromosomes causes sex differences in adiposity in mice. PLoS Genet 8:e1002709. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pgen.1002709&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22589744&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 51. Chi L, Gao B, Bian X, Tu P, Ru H, and Lu K (2017) Manganese-induced sex-specific gut microbiome perturbations in C57BL/6 mice. Toxicol Appl Pharmacol 331:142–153. 52. Chong PL, Laight D, Aspinall RJ, Higginson A, and Cummings MH (2021) A randomised placebo controlled trial of VSL#3® probiotic on biomarkers of cardiovascular risk and liver injury in non-alcoholic fatty liver disease. BMC Gastroenterol 21:144. 53. Chriett S, Dąbek A, Wojtala M, Vidal H, Balcerczyk A, and Pirola L (2019) Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci Rep 9:742. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/s41598-018-36941-9&link_type=DOI) 54. Clegg DJ, Brown LM, Woods SC, and Benoit SC (2006) Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes 55:978–987. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6ODoiNTUvNC85NzgiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 55. Clemente JC, Pehrsson EC, Blaser MJ, Sandhu K, Gao Z, Wang B, Magris M, Hidalgo G, Contreras M, Noya-Alarcón Ó, et al. (2015) The microbiome of uncontacted Amerindians. Sci Adv 1:e1500183. [FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo4OiJhZHZhbmNlcyI7czo1OiJyZXNpZCI7czoxMjoiMS8zL2UxNTAwMTgzIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 56. Clemente-Postigo M, Oliva-Olivera W, Coin-Aragüez L, Ramos-Molina B, Giraldez-Perez RM, Lhamyani S, Alcaide-Torres J, Perez-Martinez P, El Bekay R, Cardona F, et al. (2019) Metabolic endotoxemia promotes adipose dysfunction and inflammation in human obesity. Am J Physiol Endocrinol Metab 316:E319–E332. 57. Collado MC, Isolauri E, Laitinen K, and Salminen S (2008) Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am J Clin Nutr 88:894–899. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpjbiI7czo1OiJyZXNpZCI7czo4OiI4OC80Lzg5NCI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 58. Collins FL, Rios-Arce ND, Atkinson S, Bierhalter H, Schoenherr D, Bazil JN, McCabe LR, and Parameswaran N (2017) Temporal and regional intestinal changes in permeability, tight junction, and cytokine gene expression following ovariectomy-induced estrogen deficiency. Physiol Rep 5:e13263. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InBoeXNyZXBvcnRzIjtzOjU6InJlc2lkIjtzOjEwOiI1LzkvZTEzMjYzIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 59. Costabile A, Buttarazzi I, Kolida S, Quercia S, Baldini J, Swann JR, Brigidi P, and Gibson GR (2017) An in vivo assessment of the cholesterol-lowering efficacy of Lactobacillus plantarum ECGC 13110402 in normal to mildly hypercholesterolaemic adults. PLoS One 12:e0187964. 60. Craven L, Rahman A, Nair Parvathy S, Beaton M, Silverman J, Qumosani K, Hramiak I, Hegele R, Joy T, Meddings J, et al. (2020) Allogenic fecal microbiota transplantation in patients with nonalcoholic fatty liver disease improves abnormal small intestinal permeability: a randomized control trial. Am J Gastroenterol 115:1055–1065. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 61. Dalile B, Vervliet B, Bergonzelli G, Verbeke K, and Van Oudenhove L (2020) Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: a randomized, placebo-controlled trial. Neuropsychopharmacology 45:2257–2266. 62. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505:559–563. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature12820&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24336217&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000329995000042&link_type=ISI) 63. de la Cuesta-Zuluaga J, Kelley ST, Chen Y, Escobar JS, Mueller NT, Ley RE, McDonald D, Huang S, Swafford AD, Knight R, et al. (2019) Age- and sex-dependent patterns of gut microbial diversity in human adults. mSystems 4:e00261-19. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoibXN5cyI7czo1OiJyZXNpZCI7czoxMzoiNC80L2UwMDI2MS0xOSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 64. de la Cuesta-Zuluaga J, Mueller NT, Álvarez-Quintero R, Velásquez-Mejía EP, Sierra JA, Corrales-Agudelo V, Carmona JA, Abad JM, and Escobar JS (2018) Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk factors. Nutrients 11: 51. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.3390/nu11010051&link_type=DOI) 65. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, and Bakker BM (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54:2325–2340. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjk6IjU0LzkvMjMyNSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 66. de Rooij SR, Nijpels G, Nilsson PM, Nolan JJ, Gabriel R, Bobbioni-Harsch E, Mingrone G, and Dekker JM (2009) Low-grade chronic inflammation in the relationship between insulin sensitivity and cardiovascular disease (RISC) population: associations with insulin resistance and cardiometabolic risk profile. Diabetes Care 32:1295–1301. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czo5OiIzMi83LzEyOTUiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 67. Diebel ME, Diebel LN, Manke CW, and Liberati DM (2015) Estrogen modulates intestinal mucus physiochemical properties and protects against oxidant injury. J Trauma Acute Care Surg 78:94–99. 68. Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB, and Ahn J (2015) Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS One 10:e0124599. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0124599&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25874569&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 69. Durgan DJ, Ganesh BP, Cope JL, Ajami NJ, Phillips SC, Petrosino JF, Hollister EB, and Bryan RM Jr. (2016) Role of the gut microbiome in obstructive sleep apnea-induced hypertension. Hypertension 67:469–474. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1161/HYPERTENSIONAHA.115.06672&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 70. Elderman M, de Vos P, and Faas M (2018) Role of microbiota in sexually dimorphic immunity. Front Immunol 9:1018. 71. Elderman M, Sovran B, Hugenholtz F, Graversen K, Huijskes M, Houtsma E, Belzer C, Boekschoten M, de Vos P, Dekker J, et al. (2017) The effect of age on the intestinal mucus thickness, microbiota composition and immunity in relation to sex in mice. PLoS One 12:e0184274. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0184274&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=28898292&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 72. Elkhatib MAW, Mroueh A, Rafeh RW, Sleiman F, Fouad H, Saad EI, Fouda MA, Elgaddar O, Issa K, Eid AH, et al. (2019) Amelioration of perivascular adipose inflammation reverses vascular dysfunction in a model of nonobese prediabetic metabolic challenge: potential role of antidiabetic drugs. Transl Res 214:121–143. 73. Esmaeilinezhad Z, Barati-Boldaji R, Brett NR, de Zepetnek JOT, Bellissimo N, Babajafari S, and Sohrabi Z (2020) The effect of synbiotics pomegranate juice on cardiovascular risk factors in PCOS patients: a randomized, triple-blinded, controlled trial. J Endocrinol Invest 43:539–548. 74. Felix KM, Tahsin S, and Wu HJ (2018) Host-microbiota interplay in mediating immune disorders. Ann N Y Acad Sci 1417:57–70. 75. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C, Shoelson S, et al. (2009) Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 15:930–939. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nm.2002&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19633656&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000268770400038&link_type=ISI) 76. Fleissner CK, Huebel N, Abd El-Bary MM, Loh G, Klaus S, and Blaut M (2010) Absence of intestinal microbiota does not protect mice from diet-induced obesity. Br J Nutr 104:919–929. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1017/S0007114510001303&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20441670&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 77. Freeland KR and Wolever TM (2010) Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-α. Br J Nutr 103:460–466. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1017/S0007114509991863&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19818198&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 78. Frost F, Kacprowski T, Rühlemann M, Pietzner M, Bang C, Franke A, Nauck M, Völker U, Völzke H, Dörr M, et al. (2021) Long-term instability of the intestinal microbiome is associated with metabolic liver disease, low microbiota diversity, diabetes mellitus and impaired exocrine pancreatic function. Gut 70:522–530. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjcwLzMvNTIyIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 79. Fukae J, Amasaki Y, Yamashita Y, Bohgaki T, Yasuda S, Jodo S, Atsumi T, and Koike T (2005) Butyrate suppresses tumor necrosis factor α production by regulating specific messenger RNA degradation mediated through a cis-acting AU-rich element. Arthritis Rheum 52:2697–2707. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1002/art.21258&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=16142751&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 80. Gao H, Shu Q, Chen J, Fan K, Xu P, Zhou Q, Li C, and Zheng H (2019) Antibiotic exposure has sex-dependent effects on the gut microbiota and metabolism of short-chain fatty acids and amino acids in mice. mSystems 4:e00048-19. 81. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, Cefalu WT, and Ye J (2009) Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58:1509–1517. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6OToiNTgvNy8xNTA5IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 82. Gasmi A, Mujawdiya PK, Pivina L, Doşa A, Semenova Y, Gasmi Benahmed A, and Bjørklund G (2021) Relationship between gut microbiota, gut hyperpermeability, and obesity. Curr Med Chem 28:827–839. 83. Gerdts E and Regitz-Zagrosek V (2019) Sex differences in cardiometabolic disorders. Nat Med 25:1657–1666. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/s41591-019-0643-8&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 84. Ghoshal S, Witta J, Zhong J, de Villiers W, and Eckhardt E (2009) Chylomicrons promote intestinal absorption of lipopolysaccharides. J Lipid Res 50:90–97. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjc6IjUwLzEvOTAiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 85. Gillis EE and Sullivan JC (2016) Sex differences in hypertension: recent advances. Hypertension 68:1322–1327. 86. Gollasch M (2017) Adipose-vascular coupling and potential therapeutics. Annu Rev Pharmacol Toxicol 57:417–436. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1146/annurev-pharmtox-010716-104542&link_type=DOI) 87. Grąt M, Wronka KM, Lewandowski Z, Grąt K, Krasnodębski M, Stypułkowski J, Hołówko W, Masior Ł, Kosińska I, Wasilewicz M, et al. (2017) Effects of continuous use of probiotics before liver transplantation: a randomized, double-blind, placebo-controlled trial. Clin Nutr 36:1530–1539. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 88. Greenstein AS, Khavandi K, Withers SB, Sonoyama K, Clancy O, Jeziorska M, Laing I, Yates AP, Pemberton PW, Malik RA, et al. (2009) Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients. Circulation 119:1661–1670. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjExOiIxMTkvMTIvMTY2MSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 89. Grehan MJ, Borody TJ, Leis SM, Campbell J, Mitchell H, and Wettstein A (2010) Durable alteration of the colonic microbiota by the administration of donor fecal flora. J Clin Gastroenterol 44:551–561. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/MCG.0b013e3181e5d06b&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20716985&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 90. Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, and Harrison DG (2007) Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med 204:2449–2460. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjExOiIyMDQvMTAvMjQ0OSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 91. Han G, Lee HJ, Jeong SE, Jeon CO, and Hyun S (2017) Comparative analysis of Drosophila melanogaster gut microbiota with respect to host strain, sex, and age. Microb Ecol 74:207–216. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s00248-016-0925-3&link_type=DOI) 92. Harada N, Hanada K, Minami Y, Kitakaze T, Ogata Y, Tokumoto H, Sato T, Kato S, Inui H, and Yamaji R (2020) Role of gut microbiota in sex- and diet-dependent metabolic disorders that lead to early mortality of androgen receptor-deficient male mice. Am J Physiol Endocrinol Metab 318:E525–E537. 93. Haro C, Rangel-Zúñiga OA, Alcalá-Díaz JF, Gómez-Delgado F, Pérez-Martínez P, Delgado-Lista J, Quintana-Navarro GM, Landa BB, Navas-Cortés JA, Tena-Sempere M, et al. (2016) Intestinal microbiota is influenced by gender and body mass index. PLoS One 11:e0154090. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0154090&link_type=DOI) 94. Hayashi T, Yamashita T, Watanabe H, Kami K, Yoshida N, Tabata T, Emoto T, Sasaki N, Mizoguchi T, Irino Y, et al. (2018) Gut microbiome and plasma microbiome-related metabolites in patients with decompensated and compensated heart failure. Circ J 83:182–192. 95. Heianza Y, Arase Y, Kodama S, Hsieh SD, Tsuji H, Saito K, Shimano H, Hara S, and Sone H (2013) Effect of postmenopausal status and age at menopause on type 2 diabetes and prediabetes in Japanese individuals: Toranomon Hospital Health Management Center Study 17 (TOPICS 17). Diabetes Care 36:4007–4014. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czoxMDoiMzYvMTIvNDAwNyI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 96. Heilbronn LK and Campbell LV (2008) Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des 14:1225–1230. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.2174/138161208784246153&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18473870&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000255683400009&link_type=ISI) 97. Hersoug LG, Møller P, and Loft S (2016) Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes Rev 17:297–312. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1111/obr.12370&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26712364&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 98. Hildebrandt MA, Hoffmann C, Sherrill–Mix SA, Keilbaugh SA, Hamady M, Chen YY, Knight R, Ahima RS, Bushman F, and Wu GD (2009) High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 137:1716–1724. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1053/j.gastro.2009.08.042&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19706296&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000271500700029&link_type=ISI) 99. Holzapfel WH and Schillinger U (2002) Introduction to pre- and probiotics. Food Res Int 35:109–116. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/S0963-9969(01)00171-5&link_type=DOI) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000173891100002&link_type=ISI) 100.Homma H, Hoy E, Xu DZ, Lu Q, Feinman R, and Deitch EA (2005) The female intestine is more resistant than the male intestine to gut injury and inflammation when subjected to conditions associated with shock states. Am J Physiol Gastrointest Liver Physiol 288:G466–G472. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpgi.00036.2004&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=15499084&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000226833100008&link_type=ISI) 101.Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, and Gordon JI (2001) Molecular analysis of commensal host-microbial relationships in the intestine. Science 291:881–884. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIyOTEvNTUwNS84ODEiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 102.Hotamisligil GS (2006) Inflammation and metabolic disorders. Nature 444:860–867. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature05485&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17167474&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000242805400043&link_type=ISI) 103.Huda SS, Jordan F, Bray J, Love G, Payne R, Sattar N, and Freeman DJ (2017) Visceral adipose tissue activated macrophage content and inflammatory adipokine secretion is higher in pre-eclampsia than in healthy pregnancies. Clin Sci (Lond) 131:1529–1540. 104.Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, and Vittinghoff E (1998) Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA 280:605–613. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1001/jama.280.7.605&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=9718051&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000075384300028&link_type=ISI) 105.Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, Creasy HH, Earl AM, FitzGerald MG, Fulton RS, et al; Human Microbiome Project Consortium (2012) Structure, function and diversity of the healthy human microbiome. Nature 486:207–214. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature11234&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22699609&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000305189000025&link_type=ISI) 106.Ianiro G, Tilg H, and Gasbarrini A (2016) Antibiotics as deep modulators of gut microbiota: between good and evil. Gut 65:1906–1915. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjEwOiI2NS8xMS8xOTA2IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 107.Iebba V, Totino V, Gagliardi A, Santangelo F, Cacciotti F, Trancassini M, Mancini C, Cicerone C, Corazziari E, Pantanella F, et al. (2016) Eubiosis and dysbiosis: the two sides of the microbiota. New Microbiol 39:1–12. 108.Imamov O, Morani A, Shim GJ, Omoto Y, Thulin-Andersson C, Warner M, and Gustafsson JA (2004) Estrogen receptor beta regulates epithelial cellular differentiation in the mouse ventral prostate. Proc Natl Acad Sci USA 101:9375–9380. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTAxLzI1LzkzNzUiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 109.Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, and Giardina C (2000) The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology 118:724–734. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/S0016-5085(00)70142-9&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10734024&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000086119100013&link_type=ISI) 110.Inoue D, Tsujimoto G, and Kimura I (2014) Regulation of Energy Homeostasis by GPR41. Front Endocrinol (Lausanne) 5:81. 111.Isolauri E, Salminen S, and Ouwehand AC (2004) Microbial-gut interactions in health and disease. Probiotics. Best Pract Res Clin Gastroenterol 18:299–313. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.bpg.2003.10.006&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=15123071&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 112.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. (2009) Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139:485–498. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cell.2009.09.033&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19836068&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000271259600014&link_type=ISI) 113.Ivey KL, Hodgson JM, Kerr DA, Thompson PL, Stojceski B, and Prince RL (2015) The effect of yoghurt and its probiotics on blood pressure and serum lipid profile; a randomised controlled trial. Nutr Metab Cardiovasc Dis 25:46–51. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.numecd.2014.07.012&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25171898&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 114.Jaacks LM, Vandevijvere S, Pan A, McGowan CJ, Wallace C, Imamura F, Mozaffarian D, Swinburn B, and Ezzati M (2019) The obesity transition: stages of the global epidemic. Lancet Diabetes Endocrinol 7:231–240. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/S2213-8587(19)30026-9&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 115.Jiang Z, Huang X, Huang S, Guo H, Wang L, Li X, Huang X, Wang T, Zhang L, and Sun L (2016) Sex-related differences of lipid metabolism induced by triptolide: the possible role of the LXRα/SREBP-1 signaling pathway. Front Pharmacol 7:87. 116.Joe B, McCarthy CG, Edwards JM, Cheng X, Chakraborty S, Yang T, Golonka RM, Mell B, Yeo J-Y, Bearss NR, et al. (2020) Microbiota introduced to germ-free rats restores vascular contractility and blood pressure. Hypertension 76:1847–1855. 117.Jonsson AL and Bäckhed F (2017) Role of gut microbiota in atherosclerosis. Nat Rev Cardiol 14:79–87. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nrcardio.2016.183&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 118.Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK, and Kang JX (2018) Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome 6:205. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s40168-018-0587-0&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=30424806&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 119.Kaliannan K, Wang B, Li XY, Kim KJ, and Kang JX (2015) A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci Rep 5:11276. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/srep11276&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26062993&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 120.Kallio KA, Hätönen KA, Lehto M, Salomaa V, Männistö S, and Pussinen PJ (2015) Endotoxemia, nutrition, and cardiometabolic disorders. Acta Diabetol 52:395–404. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s00592-014-0662-3&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25326898&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 121.Kappel BA and Federici M (2019) Gut microbiome and cardiometabolic risk. Rev Endocr Metab Disord 20:399–406. 122.Kasai C, Sugimoto K, Moritani I, Tanaka J, Oya Y, Inoue H, Tameda M, Shiraki K, Ito M, Takei Y, et al. (2015) Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol 15:100. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s12876-015-0330-2&link_type=DOI) 123.Kazemian N, Mahmoudi M, Halperin F, Wu JC, and Pakpour S (2020) Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome 8:36. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s40168-020-00821-0&link_type=DOI) 124.Kelly CR, Khoruts A, Staley C, Sadowsky MJ, Abd M, Alani M, Bakow B, Curran P, McKenney J, Tisch A, et al. (2016) Effect of fecal microbiota transplantation on recurrence in multiply recurrent Clostridium difficile infection: a randomized trial. Ann Intern Med 165:609–616. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.7326/M16-0271&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27547925&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 125.Khan MJ, Gerasimidis K, Edwards CA, and Shaikh MG (2016) Role of gut microbiota in the aetiology of obesity: proposed mechanisms and review of the literature. J Obes 2016:7353642. 126.Khan TJ, Ahmed YM, Zamzami MA, Mohamed SA, Khan I, Baothman OAS, Mehanna MG, and Yasir M (2018) Effect of atorvastatin on the gut microbiota of high fat diet-induced hypercholesterolemic rats. Sci Rep 8:662. 127.Khatib MA-W, Sleiman F, Saad EI, Fouad HH, Issa K, Eid A, Eid A, and El-Yazbi AF (2018) Mild hyper-caloric intake is associated with peri-vascular adipose inflammation and vascular dysfunction: modulation by antidiabetic drugs. FASEB J 32:569.511 128.Kim K-A, Gu W, Lee I-A, Joh E-H, and Kim D-H (2012) High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7:e47713. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0047713&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23091640&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 129.Kim M, Neinast MD, Frank AP, Sun K, Park J, Zehr JA, Vishvanath L, Morselli E, Amelotte M, Palmer BF, et al. (2014) ERα upregulates Phd3 to ameliorate HIF-1 induced fibrosis and inflammation in adipose tissue. Mol Metab 3:642–651. 130.Kim S, Goel R, Kumar A, Qi Y, Lobaton G, Hosaka K, Mohammed M, Handberg EM, Richards EM, Pepine CJ, et al. (2018) Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clin Sci (Lond) 132:701–718. 131.Kim SH and Reaven G (2013) Sex differences in insulin resistance and cardiovascular disease risk. J Clin Endocrinol Metab 98:E1716–E1721. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1210/jc.2013-1166&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24064694&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 132.Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, and Tsujimoto G (2011) Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci USA 108:8030–8035. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTA4LzE5LzgwMzAiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 133.Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, Terasawa K, Kashihara D, Hirano K, Tani T, et al. (2013) The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun 4:1829. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/ncomms2852&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23652017&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 134.Kitchens RL and Thompson PA (2005) Modulatory effects of sCD14 and LBP on LPS-host cell interactions. J Endotoxin Res 11:225–229. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1177/09680519050110040701&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=16176659&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000232203900005&link_type=ISI) 135.Korem T, Zeevi D, Suez J, Weinberger A, Avnit-Sagi T, Pompan-Lotan M, Matot E, Jona G, Harmelin A, Cohen N, et al. (2015) Growth dynamics of gut microbiota in health and disease inferred from single metagenomic samples. Science 349:1101–1106. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNDkvNjI1Mi8xMTAxIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 136.Koren O, Spor A, Felin J, Fåk F, Stombaugh J, Tremaroli V, Behre CJ, Knight R, Fagerberg B, Ley RE, and Bäckhed F (2011) Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci USA 108 Suppl 1:4592–4598. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoyMToiMTA4L1N1cHBsZW1lbnRfMS80NTkyIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 137.Krack A, Sharma R, Figulla HR, and Anker SD (2005) The importance of the gastrointestinal system in the pathogenesis of heart failure. Eur Heart J 26:2368–2374. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1093/eurheartj/ehi389&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=15980032&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000233044800008&link_type=ISI) 138.Lagier J-C (2014) Faecal microbiota transplantation: from practice to legislation before considering industrialization. Clin Microbiol Infect 20:1112–1118. 139.Lakka H-M, Laaksonen DE, Lakka TA, Niskanen LK, Kumpusalo E, Tuomilehto J, and Salonen JT (2002) The metabolic syndrome and total and cardiovascular disease mortality in middle-aged men. JAMA 288:2709–2716. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1001/jama.288.21.2709&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=12460094&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000179549800030&link_type=ISI) 140.Leber B, Tripolt NJ, Blattl D, Eder M, Wascher TC, Pieber TR, Stauber R, Sourij H, Oettl K, and Stadlbauer V (2012) The influence of probiotic supplementation on gut permeability in patients with metabolic syndrome: an open label, randomized pilot study. Eur J Clin Nutr 66:1110–1115. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22872030&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 141.Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto J-M, Kennedy S, et al; MetaHIT consortium (2013) Richness of human gut microbiome correlates with metabolic markers. Nature 500:541–546. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature12506&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23985870&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000323625900026&link_type=ISI) 142.Lee CH, Steiner T, Petrof EO, Smieja M, Roscoe D, Nematallah A, Weese JS, Collins S, Moayyedi P, Crowther M, et al. (2016) Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent Clostridium difficile infection: a randomized clinical trial. JAMA 315:142–149. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1001/jama.2015.18098&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26757463&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 143.Lei B, Mace B, Dawson HN, Warner DS, Laskowitz DT, and James ML (2014) Anti-inflammatory effects of progesterone in lipopolysaccharide-stimulated BV-2 microglia. PLoS One 9:e103969. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0103969&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25080336&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 144.Lemieux S, Prud’homme D, Bouchard C, Tremblay A, and Després J-P (1993) Sex differences in the relation of visceral adipose tissue accumulation to total body fatness. Am J Clin Nutr 58:463–467. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpjbiI7czo1OiJyZXNpZCI7czo4OiI1OC80LzQ2MyI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 145.Leon LJ, McCarthy FP, Direk K, Gonzalez-Izquierdo A, Prieto-Merino D, Casas JP, and Chappell L (2019) Preeclampsia and cardiovascular disease in a large UK pregnancy cohort of linked electronic health records: a CALIBER study. Circulation 140:1050–1060. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1161/CIRCULATIONAHA.118.038080&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=31545680&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 146.Leong KSW, Jayasinghe TN, Wilson BC, Derraik JGB, Albert BB, Chiavaroli V, Svirskis DM, Beck KL, Conlon CA, Jiang Y, et al. (2020) Effects of fecal microbiome transfer in adolescents with obesity: the gut bugs randomized controlled trial. JAMA Netw Open 3:e2030415. 147.Lewis CV and Taylor WR (2020) Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol 319:H1227–H1233. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpheart.00612.2020&link_type=DOI) 148.Ley RE, Turnbaugh PJ, Klein S, and Gordon JI (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444:1022–1023. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/4441022a&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17183309&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000242971100042&link_type=ISI) 149.Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, Geng B, et al. (2017) Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 5:14. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s40168-016-0222-x&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=28143587&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 150.Liang H and Ward WF (2006) PGC-1α: a key regulator of energy metabolism. Adv Physiol Educ 30:145–151. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/advan.00052.2006&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17108241&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000242047300004&link_type=ISI) 151.Lima R, Wofford M, and Reckelhoff JF (2012) Hypertension in postmenopausal women. Curr Hypertens Rep 14:254–260. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s11906-012-0260-0&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22427070&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 152.Link JC, Chen X, Arnold AP, and Reue K (2013) Metabolic impact of sex chromosomes. Adipocyte 2:74–79. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.4161/adip.23320&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23805402&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 153.Liu Y, Song X, Zhou H, Zhou X, Xia Y, Dong X, Zhong W, Tang S, Wang L, Wen S, et al. (2018) Gut microbiome associates with lipid-lowering effect of Rosuvastatin *in vivo*. Front Microbiol 9:530. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 154.Looijer-van Langen M, Hotte N, Dieleman LA, Albert E, Mulder C, and Madsen KL (2011) Estrogen receptor-β signaling modulates epithelial barrier function. Am J Physiol Gastrointest Liver Physiol 300:G621–G626. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpgi.00274.2010&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=21252046&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000289276500014&link_type=ISI) 155.Lu N, Li M, Lei H, Jiang X, Tu W, Lu Y, and Xia D (2017) Butyric acid regulates progesterone and estradiol secretion via cAMP signaling pathway in porcine granulosa cells. J Steroid Biochem Mol Biol 172:89–97. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.jsbmb.2017.06.004&link_type=DOI) 156.Lu Y-C, Yeh W-C, and Ohashi PS (2008) LPS/TLR4 signal transduction pathway. Cytokine 42:145–151. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cyto.2008.01.006&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18304834&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000256535700001&link_type=ISI) 157.Lumeng CN, Bodzin JL, and Saltiel AR (2007) Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117:175–184. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1172/JCI29881&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17200717&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000243538400022&link_type=ISI) 158.Lutsey PL, Steffen LM, and Stevens J (2008) Dietary intake and the development of the metabolic syndrome: the Atherosclerosis Risk in Communities study. Circulation 117:754–761. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjk6IjExNy82Lzc1NCI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 159.Ma G, Pan B, Chen Y, Guo C, Zhao M, Zheng L, and Chen B (2017) Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci Rep 37:BSR20160244. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InBwYmlvc2NpcmVwIjtzOjU6InJlc2lkIjtzOjE2OiIzNy8yL0JTUjIwMTYwMjQ0IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 160.Malik M, Suboc TM, Tyagi S, Salzman N, Wang J, Ying R, Tanner MJ, Kakarla M, Baker JE, and Widlansky ME (2018) Lactobacillus plantarum 299v supplementation improves vascular endothelial function and reduces inflammatory biomarkers in men with stable coronary artery disease. Circ Res 123:1091–1102. 161.Manwani B, Bentivegna K, Benashski SE, Venna VR, Xu Y, Arnold AP, and McCullough LD (2015) Sex differences in ischemic stroke sensitivity are influenced by gonadal hormones, not by sex chromosome complement. J Cereb Blood Flow Metab 35:221–229. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/jcbfm.2014.186&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25388681&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 162.Marcucci R, Valente S, Gori AM, Chiostri M, Paniccia R, Giusti B, Cau V, Lazzeri C, Gensini GF, and Abbate R (2014) Global platelet hyperreactivity and elevated C-reactive protein levels predict long term mortality in STEMI patients. Thromb Res 134:884–888. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.thromres.2014.07.020&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25135796&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 163.Mariat D, Firmesse O, Levenez F, Guimarăes V, Sokol H, Doré J, Corthier G, and Furet JP (2009) The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 9:123. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/1471-2180-9-123&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19508720&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 164.Mariño E, Richards JL, McLeod KH, Stanley D, Yap YA, Knight J, McKenzie C, Kranich J, Oliveira AC, Rossello FJ, et al. (2017) Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat Immunol 18:552–562. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/ni.3713&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=28346408&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 165.Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ, and Danska JS (2013) Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339:1084–1088. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMzkvNjEyMy8xMDg0IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 166.Martínez-Oca P, Robles-Vera I, Sánchez-Roncero A, Escrivá F, Pérez-Vizcaíno F, Duarte J, Álvarez C, and Fernández-Millán E (2020) Gut DYSBIOSIS and altered barrier function precedes the appearance of metabolic syndrome in a rat model of nutrient-induced catch-up growth. J Nutr Biochem 81:108383. 167.Martins F, Campos DH, Pagan LU, Martinez PF, Okoshi K, Okoshi MP, Padovani CR, Souza AS, Cicogna AC, and Oliveira-Junior SA (2015) High-fat diet promotes cardiac remodeling in an experimental model of obesity. Arq Bras Cardiol 105:479–486. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26291841&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 168.Matsumoto M, Kitada Y, and Naito Y (2019) Endothelial function is improved by inducing microbial polyamine production in the gut: a randomized placebo-controlled trial. Nutrients 11:1188. 169.Mayneris-Perxachs J, Arnoriaga-Rodríguez M, Luque-Córdoba D, Priego-Capote F, Pérez-Brocal V, Moya A, Burokas A, Maldonado R, and Fernández-Real J-M (2020) Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: influences of obesity and menopausal status. Microbiome 8:136. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s40168-020-00913-x&link_type=DOI) 170.Mazzini L, Mogna L, De Marchi F, Amoruso A, Pane M, Aloisio I, Cionci NB, Gaggìa F, Lucenti A, Bersano E, et al. (2018) Potential role of gut microbiota in ALS pathogenesis and possible novel therapeutic strategies. J Clin Gastroenterol 52 (Suppl 1, Proceedings from the 9th Probiotics, Prebiotics and New Foods, Nutraceuticals and Botanicals for Nutrition & Human and Microbiota Health Meeting, held in Rome, Italy from September 10 to 12, 2017):S68–S70. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/MCG.0000000000001042.&link_type=DOI) 171.Meijer RI, Bakker W, Alta C-LA, Sipkema P, Yudkin JS, Viollet B, Richter EA, Smulders YM, van Hinsbergh VW, Serné EH, et al. (2013) Perivascular adipose tissue control of insulin-induced vasoreactivity in muscle is impaired in db/db mice. Diabetes 62:590–598. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6ODoiNjIvMi81OTAiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 172.Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, Vijay-Kumar M, Pennathur S, and Joe B (2015) Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol Genomics 47:187–197. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/physiolgenomics.00136.2014&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25829393&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 173.Menon R, Watson SE, Thomas LN, Allred CD, Dabney A, Azcarate-Peril MA, and Sturino JM (2013) Diet complexity and estrogen receptor β status affect the composition of the murine intestinal microbiota. Appl Environ Microbiol 79:5763–5773. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWVtIjtzOjU6InJlc2lkIjtzOjEwOiI3OS8xOC81NzYzIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 174.Million M, Maraninchi M, Henry M, Armougom F, Richet H, Carrieri P, Valero R, Raccah D, Vialettes B, and Raoult D (2012) Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int J Obes 36:817–825. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/ijo.2011.153&link_type=DOI) 175.Misra A, Singhal N, and Khurana L (2010) Obesity, the metabolic syndrome, and type 2 diabetes in developing countries: role of dietary fats and oils. J Am Coll Nutr 29 Suppl 3)289S–301S. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1080/07315724.2010.10719844&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20823489&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000282072700006&link_type=ISI) 176.Moludi J, Maleki V, Jafari-Vayghyan H, Vaghef-Mehrabany E, and Alizadeh M (2020) Metabolic endotoxemia and cardiovascular disease: A systematic review about potential roles of prebiotics and probiotics. Clin Exp Pharmacol Physiol 47:927–939. 177.Moreira APB, Texeira TFS, Ferreira AB, Peluzio MdoC, and Alfenas RdeC (2012) Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br J Nutr 108:801–809. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1017/S0007114512001213&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22717075&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 178.Moreno-Indias I, Sánchez-Alcoholado L, Sánchez-Garrido MÁ, Martín-Núñez GM, Pérez-Jiménez F, Tena-Sempere M, Tinahones FJ, and Queipo-Ortuño MI (2016) Neonatal androgen exposure causes persistent gut microbiota dysbiosis related to metabolic disease in adult female rats. Endocrinology 157:4888–4898. 179.Morrison DJ and Preston T (2016) Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 7:189–200. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1080/19490976.2015.1134082&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26963409&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 180.Mosca L, Barrett-Connor E, and Wenger NK (2011) Sex/gender differences in cardiovascular disease prevention: what a difference a decade makes. Circulation 124:2145–2154. [FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjExOiIxMjQvMTkvMjE0NSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 181.Mueller NT, Zhang M, Juraschek SP, Miller ER, and Appel LJ (2020) Effects of high-fiber diets enriched with carbohydrate, protein, or unsaturated fat on circulating short chain fatty acids: results from the OmniHeart randomized trial. Am J Clin Nutr 111:545–554. 182.Mueller S, Saunier K, Hanisch C, Norin E, Alm L, Midtvedt T, Cresci A, Silvi S, Orpianesi C, Verdenelli MC, et al. (2006) Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Appl Environ Microbiol 72:1027–1033. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWVtIjtzOjU6InJlc2lkIjtzOjk6IjcyLzIvMTAyNyI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 183.Murphy EA, Velazquez KT, and Herbert KM (2015) Influence of high-fat diet on gut microbiota: a driving force for chronic disease risk. Curr Opin Clin Nutr Metab Care 18:515–520. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/MCO.0000000000000209&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26154278&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 184.Naruszewicz M, Johansson M-L, Zapolska-Downar D, and Bukowska H (2002) Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers. Am J Clin Nutr 76:1249–1255. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpjbiI7czo1OiJyZXNpZCI7czo5OiI3Ni82LzEyNDkiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 185.Nishimura S, Manabe I, and Nagai R (2009) Adipose tissue inflammation in obesity and metabolic syndrome. Discov Med 8:55–60. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19788868&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 186.Njølstad I, Arnesen E, and Lund-Larsen PG (1996) Smoking, serum lipids, blood pressure, and sex differences in myocardial infarction. A 12-year follow-up of the Finnmark Study. Circulation 93:450–456. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjg6IjkzLzMvNDUwIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 187.Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA, and Lusis AJ (2016) Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes 7:313–322. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1080/19490976.2016.1203502&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27355107&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 188.Osibogun O, Ogunmoroti O, and Michos ED (2020) Polycystic ovary syndrome and cardiometabolic risk: opportunities for cardiovascular disease prevention. Trends Cardiovasc Med 30:399–404. 189.Ott SJ, El Mokhtari NE, Musfeldt M, Hellmig S, Freitag S, Rehman A, Kühbacher T, Nikolaus S, Namsolleck P, Blaut M, et al. (2006) Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation 113:929–937. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjk6IjExMy83LzkyOSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 190.Ott SJ, Waetzig GH, Rehman A, Moltzau-Anderson J, Bharti R, Grasis JA, Cassidy L, Tholey A, Fickenscher H, Seegert D, et al. (2017) Efficacy of sterile fecal filtrate transfer for treating patients with clostridium difficile infection. Gastroenterology 152:799–811.e7. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1053/j.gastro.2016.11.010&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27866880&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 191.Palmer BF and Clegg DJ (2014) Oxygen sensing and metabolic homeostasis. Mol Cell Endocrinol 397:51–58. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.mce.2014.08.001&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25132648&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 192.Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BA, Forslund K, Hildebrand F, Prifti E, Falony G, et al; MetaHIT Consortium (2016) Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535:376–381. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature18646&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27409811&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 193.Pei J, Harakalova M, den Ruijter H, Pasterkamp G, Duncker DJ, Verhaar MC, Asselbergs FW, and Cheng C (2017) Cardiorenal disease connection during post-menopause: the protective role of estrogen in uremic toxins induced microvascular dysfunction. Int J Cardiol 238:22–30. 194.Peng C, Xu X, Li Y, Li X, Yang X, Chen H, Zhu Y, Lu N, and He C (2020) Sex-specific association between the gut microbiome and high-fat diet-induced metabolic disorders in mice. Biol Sex Differ 11:5. 195.Pluznick J (2014) A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes 5:202–207. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.4161/gmic.27492&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24429443&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 196.Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F, Wang T, et al. (2013) Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 110:4410–4415. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTEwLzExLzQ0MTAiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 197.Poggi M, Bastelica D, Gual P, Iglesias MA, Gremeaux T, Knauf C, Peiretti F, Verdier M, Juhan-Vague I, Tanti JF, et al. (2007) C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia 50:1267–1276. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s00125-007-0654-8&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17426960&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000246271600020&link_type=ISI) 198.Poll BG, Cheema MU, and Pluznick JL (2020) Gut microbial metabolites and blood pressure regulation: focus on SCFAs and TMAO. Physiology (Bethesda) 35:275–284. 199.Pradhan AD (2014) Sex differences in the metabolic syndrome: implications for cardiovascular health in women. Clin Chem 60:44–52. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2xpbmNoZW0iO3M6NToicmVzaWQiO3M6NzoiNjAvMS80NCI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 200.Putignani L, Del Chierico F, Petrucca A, Vernocchi P, and Dallapiccola B (2014) The human gut microbiota: a dynamic interplay with the host from birth to senescence settled during childhood. Pediatr Res 76:2–10. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/pr.2014.49&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24732106&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000337663900001&link_type=ISI) 201.Qi Y, Aranda JM, Rodriguez V, Raizada MK, and Pepine CJ (2015) Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension—a case report. Int J Cardiol 201:157–158. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.ijcard.2015.07.078&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26301638&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 202.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464:59–65. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature08821&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20203603&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000275117500033&link_type=ISI) 203.Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, et al. (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490:55–60. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature11450&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23023125&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000309446800031&link_type=ISI) 204.Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, Guo J, Le Chatelier E, Yao J, Wu L, et al. (2014) Alterations of the human gut microbiome in liver cirrhosis. Nature 513:59–64. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature13568&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25079328&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 205.Rabot S, Membrez M, Bruneau A, Gérard P, Harach T, Moser M, Raymond F, Mansourian R, and Chou CJ (2010) Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J 24:4948–4959. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1096/fj.10-164921&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20724524&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000284824400033&link_type=ISI) 206.Rafeh R, Viveiros A, Oudit GY, and El-Yazbi AF (2020) Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 134:827–851. 207.Rahat-Rozenbloom S, Fernandes J, Gloor GB, and Wolever TM (2014) Evidence for greater production of colonic short-chain fatty acids in overweight than lean humans. Int J Obes 38:1525–1531. 208.Razavi AC, Potts KS, Kelly TN, and Bazzano LA (2019) Sex, gut microbiome, and cardiovascular disease risk. Biol Sex Differ 10:29. 209.Reijnders D, Goossens GH, Hermes GD, Neis EP, van der Beek CM, Most J, Holst JJ, Lenaerts K, Kootte RS, Nieuwdorp M, et al. (2016) Effects of gut microbiota manipulation by antibiotics on host metabolism in obese humans: a randomized double-blind placebo-controlled trial. Cell Metab 24:63–74. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cmet.2016.06.016&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27411009&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 210.Restini CBA, Fink GD, and Watts SW (2021) Vascular reactivity stimulated by TMA and TMAO: are perivascular adipose tissue and endothelium involved? Pharmacol Res 163:105273. 211.Rettew JA, Huet YM, and Marriott I (2009) Estrogens augment cell surface TLR4 expression on murine macrophages and regulate sepsis susceptibility in vivo. Endocrinology 150:3877–3884. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1210/en.2009-0098&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19406943&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000268158400054&link_type=ISI) 212.Rettew JA, Huet-Hudson YM, and Marriott I (2008) Testosterone reduces macrophage expression in the mouse of toll-like receptor 4, a trigger for inflammation and innate immunity. Biol Reprod 78:432–437. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1095/biolreprod.107.063545&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18003947&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000253352700006&link_type=ISI) 213.Rezazadeh L, Gargari BP, Jafarabadi MA, and Alipour B (2019) Effects of probiotic yogurt on glycemic indexes and endothelial dysfunction markers in patients with metabolic syndrome. Nutrition 62:162–168. 214.Ribas V, Nguyen MTA, Henstridge DC, Nguyen A-K, Beaven SW, Watt MJ, and Hevener AL (2010) Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERα-deficient mice. Am J Physiol Endocrinol Metab 298:E304–E319. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpendo.00504.2009&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19920214&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000273695500018&link_type=ISI) 215.Robles-Vera I, Toral M, de la Visitación N, Sánchez M, Gómez-Guzmán M, Muñoz R, Algieri F, Vezza T, Jiménez R, Gálvez J, et al. (2020) Changes to the gut microbiota induced by losartan contributes to its antihypertensive effects. Br J Pharmacol 177:2006–2023. 216.Rocha DM, Caldas AP, Oliveira LL, Bressan J, and Hermsdorff HH (2016) Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis 244:211–215. 217.Rodriguez J, Olivares M, and Delzenne NM (2020) Implication of the gut microbiota in metabolic inflammation associated with nutritional disorders and obesity. Mol Nutr Food Res 65:1900481. 218.Romão da Silva LF, de Oliveira Y, de Souza EL, de Luna Freire MO, Braga VA, Magnani M, and de Brito Alves JL (2020) Effects of probiotic therapy on cardio-metabolic parameters and autonomic modulation in hypertensive women: a randomized, triple-blind, placebo-controlled trial. Food Funct 11:7152–7163. 219.Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, et al; Writing Group for the Women’s Health Initiative Investigators (2002) Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA 288:321–333. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1001/jama.288.3.321&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=12117397&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000176807000024&link_type=ISI) 220.Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, Costea PI, Godneva A, Kalka IN, Bar N, et al. (2018) Environment dominates over host genetics in shaping human gut microbiota. Nature 555:210–215. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature25973&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=29489753&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 221.Ruscica M, Pavanello C, Gandini S, Macchi C, Botta M, Dall’Orto D, Del Puppo M, Bertolotti M, Bosisio R, and Mombelli G (2019) Nutraceutical approach for the management of cardiovascular risk—a combination containing the probiotic Bifidobacterium longum BB536 and red yeast rice extract: results from a randomized, double-blind, placebo-controlled study. Nutr J 18:1–9. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s12937-019-0450-6&link_type=DOI) 222.Saad MJ, Santos A, and Prada PO (2016) Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda) 31:283–293. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/physiol.00041.2015&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27252163&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 223.Säemann MD, Böhmig GA, Österreicher CH, Burtscher H, Parolini O, Diakos C, Stöckl J, Hörl WH, and Zlabinger GJ (2000) Anti-inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL-12 and up-regulation of IL-10 production. FASEB J 14:2380–2382. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=11024006&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000165723400004&link_type=ISI) 224.Saha S, Tariq R, Tosh PK, Pardi DS, and Khanna S (2019) Faecal microbiota transplantation for eradicating carriage of multidrug-resistant organisms: a systematic review. Clin Microbiol Infect 25:958–963. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 225.Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, et al. (2008) Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 105:16767–16772. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTA1LzQzLzE2NzY3IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 226.Sandek A, Bjarnason I, Volk HD, Crane R, Meddings JB, Niebauer J, Kalra PR, Buhner S, Herrmann R, Springer J, et al. (2012) Studies on bacterial endotoxin and intestinal absorption function in patients with chronic heart failure. Int J Cardiol 157:80–85. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.ijcard.2010.12.016&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=21190739&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 227.Santisteban MM, Qi Y, Zubcevic J, Kim S, Yang T, Shenoy V, Cole-Jeffrey CT, Lobaton GO, Stewart DC, Rubiano A, et al. (2017) Hypertension-linked pathophysiological alterations in the gut. Circ Res 120:312–323. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6OToiMTIwLzIvMzEyIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 228.Santos-Marcos JA, Barroso A, Rangel-Zuñiga OA, Perdices-Lopez C, Haro C, Sanchez-Garrido MA, Molina-Abril H, Ohlsson C, Perez-Martinez P, Poutanen M, et al. (2020) Interplay between gonadal hormones and postnatal overfeeding in defining sex-dependent differences in gut microbiota architecture. Aging (Albany NY) 12:19979–20000. 229.Santos-Marcos JA, Haro C, Vega-Rojas A, Alcala-Diaz JF, Molina-Abril H, Leon-Acuña A, Lopez-Moreno J, Landa BB, Tena-Sempere M, Perez-Martinez P, et al. (2019) Sex differences in the gut microbiota as potential determinants of gender predisposition to disease. Mol Nutr Food Res 63:e1800870. 230.Santos-Marcos JA, Rangel-Zuñiga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, Landa BB, Tena-Sempere M, Perez-Martinez P, Lopez-Miranda J, et al. (2018) Influence of gender and menopausal status on gut microbiota. Maturitas 116:43–53. 231.Satokari R (2020) High intake of sugar and the balance between pro- and anti-inflammatory gut bacteria. Nutrients 12:1348. 232.Saxton SN, Clark BJ, Withers SB, Eringa EC, and Heagerty AM (2019) Mechanistic links between obesity, diabetes, and blood pressure: role of perivascular adipose tissue. Physiol Rev 99:1701–1763. 233.Schepici G, Silvestro S, Bramanti P, and Mazzon E (2019) The gut microbiota in multiple sclerosis: an overview of clinical trials. Cell Transplant 28:1507–1527. 234.Schugar RC, Shih DM, Warrier M, Helsley RN, Burrows A, Ferguson D, Brown AL, Gromovsky AD, Heine M, Chatterjee A, et al. (2017) The TMAO-producing enzyme flavin-containing monooxygenase 3 regulates obesity and the beiging of white adipose tissue. Cell Rep 19:2451–2461. 235.Schunkert H, Danser AH, Hense HW, Derkx FH, Kürzinger S, and Riegger GA (1997) Effects of estrogen replacement therapy on the renin-angiotensin system in postmenopausal women. Circulation 95:39–45. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjc6Ijk1LzEvMzkiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 236.Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, and Hardt PD (2010) Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 18:190–195. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/oby.2009.167&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=19498350&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 237.Scorletti E, Afolabi PR, Miles EA, Smith DE, Almehmadi A, Alshathry A, Moyses HE, Clough GF, Wright M, Patel J. et al. (2018) Design and rationale of the INSYTE study: a randomised, placebo controlled study to test the efficacy of a synbiotic on liver fat, disease biomarkers and intestinal microbiota in non-alcoholic fatty liver disease. Contemp Clin Trials 71:113–123. 238.Segain JP, Raingeard de la Blétière D, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blottière HM, and Galmiche JP (2000) Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease. Gut 47:397–403. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjQ3LzMvMzk3IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 239.Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, Lusis AJ, and Shih DM (2016) Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J Am Heart Assoc 5:e002767. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NToiYWhhb2EiO3M6NToicmVzaWQiO3M6MTE6IjUvMi9lMDAyNzY3IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 240.Sen T, Cawthon CR, Ihde BT, Hajnal A, DiLorenzo PM, de La Serre CB, and Czaja K (2017) Diet-driven microbiota dysbiosis is associated with vagal remodeling and obesity. Physiol Behav 173:305–317. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.physbeh.2017.02.027&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 241.Serino M, Luche E, Gres S, Baylac A, Bergé M, Cenac C, Waget A, Klopp P, Iacovoni J, Klopp C, et al. (2012) Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 61:543–553. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjYxLzQvNTQzIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 242.Severance EG, Gressitt KL, Stallings CR, Katsafanas E, Schweinfurth LA, Savage CLG, Adamos MB, Sweeney KM, Origoni AE, Khushalani S, et al. (2017) Probiotic normalization of Candida albicans in schizophrenia: a randomized, placebo-controlled, longitudinal pilot study. Brain Behav Immun 62:41–45. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 243.Shah A, Mehta N, and Reilly MP (2008) Adipose inflammation, insulin resistance, and cardiovascular disease. JPEN J Parenter Enteral Nutr 32:638–644. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1177/0148607108325251&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=18974244&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000260446800009&link_type=ISI) 244.Shastri P, McCarville J, Kalmokoff M, Brooks SPJ, and Green-Johnson JM (2015) Sex differences in gut fermentation and immune parameters in rats fed an oligofructose-supplemented diet. Biol Sex Differ 6:13. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/s13293-015-0031-0&link_type=DOI) 245.Sheth SU, Lu Q, Twelker K, Sharpe SM, Qin X, Reino DC, Lee MA, Xu DZ, and Deitch EA (2010) Intestinal mucus layer preservation in female rats attenuates gut injury after trauma-hemorrhagic shock. J Trauma 68:279–288. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1097/TA.0b013e3181caa6bd&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=20154538&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 246.Shi H, Zhang B, Abo-Hamzy T, Nelson JW, Ambati CSR, Petrosino JF, Bryan RM Jr., and Durgan DJ (2021) Restructuring the gut microbiota by intermittent fasting lowers blood pressure. Circ Res 128:1240–1254. 247.Smits LP, Kootte RS, Levin E, Prodan A, Fuentes S, Zoetendal EG, Wang Z, Levison BS, Cleophas MCP, Kemper EM, et al. (2018) Effect of vegan fecal microbiota transplantation on carnitine‐ and choline‐derived trimethylamine‐N‐oxide production and vascular inflammation in patients with metabolic syndrome. J Am Heart Assoc 7:e008342. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NToiYWhhb2EiO3M6NToicmVzaWQiO3M6MTE6IjcvNy9lMDA4MzQyIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 248.Smits SA, Leach J, Sonnenburg ED, Gonzalez CG, Lichtman JS, Reid G, Knight R, Manjurano A, Changalucha J, Elias JE, et al. (2017) Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 357:802–806. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTcvNjM1My84MDIiO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 249.Sobrino A, Vallejo S, Novella S, Lázaro-Franco M, Mompeón A, Bueno-Betí C, Walther T, Sánchez-Ferrer C, Peiró C, and Hermenegildo C (2017) Mas receptor is involved in the estrogen-receptor induced nitric oxide-dependent vasorelaxation. Biochem Pharmacol 129:67–72. 250.Song C-H, Kim N, Nam RH, Choi SI, Lee H-N, and Surh Y-J (2020) 17β-Estradiol supplementation changes gut microbiota diversity in intact and colorectal cancer-induced ICR male mice. Sci Rep 10:12283. 251.Stadlbauer V, Leber B, Lemesch S, Trajanoski S, Bashir M, Horvath A, Tawdrous M, Stojakovic T, Fauler G, Fickert P, et al. (2015) Lactobacillus casei Shirota supplementation does not restore gut microbiota composition and gut barrier in metabolic syndrome: a randomized pilot study. PLoS One 10:e0141399. 252.Stefan N (2020) Metabolically healthy and unhealthy normal weight and obesity. Endocrinol Metab (Seoul) 35:487–493. 253.Stoll LL, Denning GM, and Weintraub NL (2004) Potential role of endotoxin as a proinflammatory mediator of atherosclerosis. Arterioscler Thromb Vasc Biol 24:2227–2236. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYXR2YmFoYSI7czo1OiJyZXNpZCI7czoxMDoiMjQvMTIvMjIyNyI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 254.Szulińska M, Łoniewski I, van Hemert S, Sobieska M, and Bogdański P (2018) Dose-dependent effects of multispecies probiotic supplementation on the lipopolysaccharide (LPS) level and cardiometabolic profile in obese postmenopausal women: a 12-week randomized clinical trial. Nutrients 10:773. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 255.Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, Wu Y, and Hazen SL (2014) Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 64:1908–1914. [FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjEwOiI2NC8xOC8xOTA4IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 256.Tang WHW, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li XS, Levison BS, and Hazen SL (2015) Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res 116:448–455. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6OToiMTE2LzMvNDQ4IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 257.Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, and Hazen SL (2013) Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 368:1575–1584. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1056/NEJMoa1109400&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23614584&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000317956300006&link_type=ISI) 258.Tang WHW, Wang Z, Li XS, Fan Y, Li DS, Wu Y, and Hazen SL (2017) Increased trimethylamine N-oxide portends high mortality risk independent of glycemic control in tatients with type 2 diabetes mellitus. Clin Chem 63:297–306. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2xpbmNoZW0iO3M6NToicmVzaWQiO3M6ODoiNjMvMS8yOTciO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 259.Taylor LE and Sullivan JC (2016) Sex differences in obesity-induced hypertension and vascular dysfunction: a protective role for estrogen in adipose tissue inflammation? Am J Physiol Regul Integr Comp Physiol 311:R714–R720. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1152/ajpregu.00202.2016&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=27511280&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 260.Taylor WR and Takemiya K (2017) Hypertension opens the flood gates to the gut microbiota. Circ Res 120:249–251. [FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6OToiMTIwLzIvMjQ5IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 261.Tedelind S, Westberg F, Kjerrulf M, and Vidal A (2007) Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol 13:2826–2832. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.3748/wjg.v13.i20.2826&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17569118&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000247325000008&link_type=ISI) 262.Teixeira TF, Grześkowiak Ł, Franceschini SC, Bressan J, Ferreira CL, and Peluzio MC (2013) Higher level of faecal SCFA in women correlates with metabolic syndrome risk factors. Br J Nutr 109:914–919. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1017/S0007114512002723&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23200109&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 263.Tenorio-Jiménez C, Martínez-Ramírez MJ, Del Castillo-Codes I, Arraiza-Irigoyen C, Tercero-Lozano M, Camacho J, Chueca N, García F, Olza J, Plaza-Díaz J, et al. (2019) Lactobacillus reuteri V3401 reduces inflammatory biomarkers and modifies the gastrointestinal microbiome in adults with metabolic syndrome: the PROSIR study. Nutrients 11:1761. 264.ter Horst R, van den Munckhof ICL, Schraa K, Aguirre-Gamboa R, Jaeger M, Smeekens SP, Brand T, Lemmers H, Dijkstra H, Galesloot TE, et al. (2020) Sex-specific regulation of inflammation and metabolic syndrome in obesity. Arterioscler Thromb Vasc Biol 40:1787–1800. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1161/ATVBAHA.120.314508&link_type=DOI) 265.Tilg H, Zmora N, Adolph TE, and Elinav E (2019) The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 20:40–54. 266.Toral M, Robles-Vera I, de la Visitación N, Romero M, Yang T, Sánchez M, Gómez-Guzmán M, Jiménez R, Raizada MK, and Duarte J (2019) Critical role of the interaction gut microbiota—sympathetic nervous system in the regulation of blood pressure. Front Physiol 10:231. 267.Traglia M, Bseiso D, Gusev A, Adviento B, Park DS, Mefford JA, Zaitlen N, and Weiss LA (2017) Genetic mechanisms leading to sex differences across common diseases and anthropometric traits. Genetics 205:979–992. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZ2VuZXRpY3MiO3M6NToicmVzaWQiO3M6OToiMjA1LzIvOTc5IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 268.Trimigno A, Khakimov B, Mejia JLC, Mikkelsen MS, Kristensen M, Jespersen BM, and Engelsen SB (2017) Identification of weak and gender specific effects in a short 3 weeks intervention study using barley and oat mixed linkage β-glucan dietary supplements: a human fecal metabolome study by GC-MS. Metabolomics 13:108. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1007/s11306-017-1247-2&link_type=DOI) 269.Trip MD, Cats VM, van Capelle FJ, and Vreeken J (1990) Platelet hyperreactivity and prognosis in survivors of myocardial infarction. N Engl J Med 322:1549–1554. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1056/NEJM199005313222201&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=2336086&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=A1990DF14000001&link_type=ISI) 270.Trøseid M, Nestvold TK, Rudi K, Thoresen H, Nielsen EW, and Lappegård KT (2013) Plasma lipopolysaccharide is closely associated with glycemic control and abdominal obesity: evidence from bariatric surgery. Diabetes Care 36:3627–3632. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czoxMDoiMzYvMTEvMzYyNyI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 271.Trotter RE, Vazquez AR, Grubb DS, Freedman KE, Grabos LE, Jones S, Gentile CL, Melby CL, Johnson SA, and Weir TL (2020) *Bacillus subtilis* DE111 intake may improve blood lipids and endothelial function in healthy adults. Benef Microbes 11:621–630. 272.Tukiainen T, Pirinen M, Sarin AP, Ladenvall C, Kettunen J, Lehtimäki T, Lokki ML, Perola M, Sinisalo J, Vlachopoulou E, et al. (2014) Chromosome X-wide association study identifies Loci for fasting insulin and height and evidence for incomplete dosage compensation. PLoS Genet 10:e1004127. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pgen.1004127&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24516404&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 273.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, and Gordon JI (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444:1027–1031. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature05414&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=17183312&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000242971100045&link_type=ISI) 274.Tuteja S and Ferguson JF (2019) Gut microbiome and response to cardiovascular drugs. Circ Genom Precis Med 12:421–429. 275.Tyakht AV, Kostryukova ES, Popenko AS, Belenikin MS, Pavlenko AV, Larin AK, Karpova IY, Selezneva OV, Semashko TA, Ospanova EA, et al. (2013) Human gut microbiota community structures in urban and rural populations in Russia. Nat Commun 4:2469. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=24036685&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 276.van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, Kuijper EJ, Bartelsman JF, Tijssen JG, et al. (2013) Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 368:407–415. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1056/NEJMoa1205037&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23323867&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000314215400005&link_type=ISI) 277.Velagapudi VR, Hezaveh R, Reigstad CS, Gopalacharyulu P, Yetukuri L, Islam S, Felin J, Perkins R, Borén J, Oresic M, et al. (2010) The gut microbiota modulates host energy and lipid metabolism in mice. J Lipid Res 51:1101–1112. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjk6IjUxLzUvMTEwMSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 278.Vidali G, Boffa LC, Mann RS, and Allfrey VG (1978) Reversible effects of Na-butyrate on histone acetylation. Biochem Biophys Res Commun 82:223–227. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/0006-291X(78)90599-5&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=666835&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=A1978EZ76400033&link_type=ISI) 279.von Bibra H, Paulus W, and St John Sutton M (2016) Cardiometabolic syndrome and increased risk of heart failure. Curr Heart Fail Rep 13:219–229. 280.Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JFWM, Dallinga–Thie GM, Ackermans MT, Serlie MJ, Oozeer R, et al. (2012) Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143: 913–916. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1053/j.gastro.2012.06.031&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22728514&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000309361800022&link_type=ISI) 281.Wada-Hiraike O, Imamov O, Hiraike H, Hultenby K, Schwend T, Omoto Y, Warner M, and Gustafsson JA (2006) Role of estrogen receptor beta in colonic epithelium. Proc Natl Acad Sci USA 103:2959–2964. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTAzLzgvMjk1OSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 282.Wang D, Wang C, Wu X, Zheng W, Sandberg K, Ji H, Welch WJ, and Wilcox CS (2014) Endothelial dysfunction and enhanced contractility in microvessels from ovariectomized rats: roles of oxidative stress and perivascular adipose tissue. Hypertension 63:1063–1069. 283.Wang S, Xu M, Wang W, Cao X, Piao M, Khan S, Yan F, Cao H, and Wang B (2016) Systematic review: adverse events of fecal microbiota transplantation. PLoS One 11:e0161174. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1371/journal.pone.0161174&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 284.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, et al. (2011) Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472:57–63. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature09922&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=21475195&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000289199400036&link_type=ISI) 285.Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, et al. (2015) Non-lethal inhibition of gut microbial yrimethylamine production for the treatment of atherosclerosis. Cell 163:1585–1595. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cell.2015.11.055&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26687352&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 286.Wensveen FM, Valentić S, Šestan M, Turk Wensveen T, and Polić B (2015) The “big bang” in obese fat: events initiating obesity-induced adipose tissue inflammation. Eur J Immunol 45:2446–2456. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1002/eji.201545502&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26220361&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 287.Wenzel U, Turner JE, Krebs C, Kurts C, Harrison DG, and Ehmke H (2016) Immune mechanisms in arterial hypertension. J Am Soc Nephrol 27:677–686. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiam5lcGhyb2wiO3M6NToicmVzaWQiO3M6ODoiMjcvMy82NzciO3M6NDoiYXRvbSI7czoyMzoiL21vbHBoYXJtLzEwMi8xLzYwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 288.Wexler HM (2007) Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev 20:593–621. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY21yIjtzOjU6InJlc2lkIjtzOjg6IjIwLzQvNTkzIjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 289.WHO (2017) Cardiovascular diseases (CVDs) fact sheet, World Health Organization, Switzerland. 290.Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, Haase S, Mähler A, Balogh A, Markó L, et al. (2017) Salt-responsive gut commensal modulates TH17 axis and disease. Nature 551:585–589. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nature24628&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 291.Williams NT (2010) Probiotics. Am J Health Syst Pharm 67:449–458. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpocCI7czo1OiJyZXNpZCI7czo4OiI2Ny82LzQ0OSI7czo0OiJhdG9tIjtzOjIzOiIvbW9scGhhcm0vMTAyLzEvNjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 292.Wu KL, Chan SH, and Chan JY (2012) Neuroinflammation and oxidative stress in rostral ventrolateral medulla contribute to neurogenic hypertension induced by systemic inflammation. J Neuroinflammation 9:212. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1186/1742-2094-9-212&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=22958438&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 293.Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong M, Qi Y, Zubcevic J, et al. (2015) Gut dysbiosis is linked to hypertension. Hypertension 65:1331–1340. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1161/HYPERTENSIONAHA.115.05315&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=25870193&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 294.Yuan X, Chen R, Zhang Y, Lin X, and Yang X (2020) Sexual dimorphism of gut microbiota at different pubertal status. Microb Cell Fact 19:152. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 295.Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, and Chervonsky AV (2013) Gender bias in autoimmunity is influenced by microbiota. Immunity 39:400–412. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.immuni.2013.08.013&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=23973225&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) [Web of Science](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=000330948800020&link_type=ISI) 296.Zeevi D, Korem T, Godneva A, Bar N, Kurilshikov A, Lotan-Pompan M, Weinberger A, Fu J, Wijmenga C, Zhernakova A, et al. (2019) Structural variation in the gut microbiome associates with host health. Nature 568:43–48. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/s41586-019-1065-y&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=30918406&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 297.Zhang Z, Mocanu V, Cai C, Dang J, Slater L, Deehan EC, Walter J, and Madsen KL (2019b) Impact of fecal microbiota transplantation on obesity and metabolic syndrome—a systematic review. Nutrients 11:2291. 298.Zheng Y, Ley SH, and Hu FB (2018) Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol 14:88–98. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1038/nrendo.2017.151&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=29219149&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 299.Zhou Z, Bian C, Luo Z, Guille C, Ogunrinde E, Wu J, Zhao M, Fitting S, Kamen DL, Oates JC, et al. (2019) Progesterone decreases gut permeability through upregulating occludin expression in primary human gut tissues and Caco-2 cells. Sci Rep 9:8367. 300.Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, et al. (2016) Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165:111–124. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1016/j.cell.2016.02.011&link_type=DOI) [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=26972052&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom) 301.Zhuang P, Shou Q, Wang W, He L, Wang J, Chen J, Zhang Y, and Jiao J (2018) Essential fatty acids linoleic acid and α-linolenic acid sex-dependently regulate glucose homeostasis in obesity. Mol Nutr Food Res 62:e1800448. [CrossRef](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=10.1002/mnfr.201800448&link_type=DOI) 302.Zoetendal EG, Rajilić-Stojanović M, and de Vos WM (2008) High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut 57:1605–1615. [Abstract/FREE Full Text](http://molpharm.aspetjournals.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjEwOiI1Ny8xMS8xNjA1IjtzOjQ6ImF0b20iO3M6MjM6Ii9tb2xwaGFybS8xMDIvMS82MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 303.Zore T, Palafox M, and Reue K (2018) Sex differences in obesity, lipid metabolism, and inflammation—a role for the sex chromosomes? Mol Metab 15:35–44. [PubMed](http://molpharm.aspetjournals.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmolpharm%2F102%2F1%2F60.atom)